首页 > 最新文献

Translational lung cancer research最新文献

英文 中文
The prognostic importance of the pan-immune-inflammation value (PIV) in lung cancer: a systematic review and meta-analysis. 泛免疫炎症值(PIV)在肺癌预后中的重要性:一项系统综述和荟萃分析。
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-10-31 Epub Date: 2025-10-29 DOI: 10.21037/tlcr-2025-518
Guangji Cao, Quanqing Liu, Hao Wen, Yuan Zeng

Background: Preliminary studies suggest that pan-immune-inflammation value (PIV) has the potential to serve as a prognostic tool for lung cancer. However, existing studies are limited by inconsistent findings regarding the impact of high PIV on patient outcomes. To provide a more comprehensive assessment, we conducted a meta-analysis to clarify the prognostic value of PIV in lung cancer.

Methods: Two researchers independently searched the PubMed, Cochrane, Embase, and Web of Science databases for studies evaluating the associations between PIV and prognoses in lung cancer patients (up to July 15, 2025). Studies were included if they reported high versus low PIV and provided hazard ratios (HRs) with 95% confidence intervals (CIs) for overall survival (OS), progression-free survival (PFS), etc. Study quality was assessed using the Newcastle-Ottawa Scale (NOS). Pooled HRs and 95% CIs were calculated to determine the associations between PIV and patient prognosis.

Results: A total of ten studies comprising 1,969 patients were included. Meta-analyses demonstrated that high PIV was significantly associated with OS (HR =2.86, 95% CI: 2.23-3.65, P<0.001) and PFS (HR =2.06, 95% CI: 1.65-2.59, P<0.001) in lung cancer patients. In non-small cell lung cancer (NSCLC) patients, high PIV was significantly associated with worse OS (HR =2.76, 95% CI: 2.14-3.56, P<0.001) and PFS (HR =1.94, 95% CI: 1.55-2.42, P<0.001). In small cell lung cancer (SCLC) patients, even stronger associations were observed for OS (HR =3.47, 95% CI: 2.21-5.44, P<0.001) and PFS (HR =2.33, 95% CI: 1.63-3.33, P<0.001). Subgroup analyses further confirmed that PIV served as a critical prognostic marker for both OS and PFS. All studies were of high quality according to the NOS.

Conclusions: PIV can serve as an independent prognostic biomarker for survival outcomes in lung cancer patients. Therefore, incorporating PIV into prognostic assessments may provide additional support for individualized treatment decision-making.

背景:初步研究表明,泛免疫炎症值(PIV)有可能作为肺癌的预后工具。然而,现有的研究受到关于高PIV对患者预后影响的不一致发现的限制。为了提供更全面的评估,我们进行了一项荟萃分析,以阐明PIV在肺癌中的预后价值。方法:两位研究人员独立检索了PubMed、Cochrane、Embase和Web of Science数据库,以评估肺癌患者PIV与预后之间的关系(截至2025年7月15日)。如果研究报告了高PIV和低PIV,并提供了总生存期(OS)、无进展生存期(PFS)的95%置信区间(ci)的风险比(hr),则纳入研究。采用纽卡斯尔-渥太华量表(NOS)评估研究质量。计算合并hr和95% ci以确定PIV与患者预后之间的关系。结果:共纳入10项研究,包括1,969例患者。荟萃分析显示,PIV高与OS显著相关(HR =2.86, 95% CI: 2.23-3.65)。结论:PIV可作为肺癌患者生存结局的独立预后生物标志物。因此,将PIV纳入预后评估可能为个性化治疗决策提供额外支持。
{"title":"The prognostic importance of the pan-immune-inflammation value (PIV) in lung cancer: a systematic review and meta-analysis.","authors":"Guangji Cao, Quanqing Liu, Hao Wen, Yuan Zeng","doi":"10.21037/tlcr-2025-518","DOIUrl":"10.21037/tlcr-2025-518","url":null,"abstract":"<p><strong>Background: </strong>Preliminary studies suggest that pan-immune-inflammation value (PIV) has the potential to serve as a prognostic tool for lung cancer. However, existing studies are limited by inconsistent findings regarding the impact of high PIV on patient outcomes. To provide a more comprehensive assessment, we conducted a meta-analysis to clarify the prognostic value of PIV in lung cancer.</p><p><strong>Methods: </strong>Two researchers independently searched the PubMed, Cochrane, Embase, and Web of Science databases for studies evaluating the associations between PIV and prognoses in lung cancer patients (up to July 15, 2025). Studies were included if they reported high versus low PIV and provided hazard ratios (HRs) with 95% confidence intervals (CIs) for overall survival (OS), progression-free survival (PFS), etc. Study quality was assessed using the Newcastle-Ottawa Scale (NOS). Pooled HRs and 95% CIs were calculated to determine the associations between PIV and patient prognosis.</p><p><strong>Results: </strong>A total of ten studies comprising 1,969 patients were included. Meta-analyses demonstrated that high PIV was significantly associated with OS (HR =2.86, 95% CI: 2.23-3.65, P<0.001) and PFS (HR =2.06, 95% CI: 1.65-2.59, P<0.001) in lung cancer patients. In non-small cell lung cancer (NSCLC) patients, high PIV was significantly associated with worse OS (HR =2.76, 95% CI: 2.14-3.56, P<0.001) and PFS (HR =1.94, 95% CI: 1.55-2.42, P<0.001). In small cell lung cancer (SCLC) patients, even stronger associations were observed for OS (HR =3.47, 95% CI: 2.21-5.44, P<0.001) and PFS (HR =2.33, 95% CI: 1.63-3.33, P<0.001). Subgroup analyses further confirmed that PIV served as a critical prognostic marker for both OS and PFS. All studies were of high quality according to the NOS.</p><p><strong>Conclusions: </strong>PIV can serve as an independent prognostic biomarker for survival outcomes in lung cancer patients. Therefore, incorporating PIV into prognostic assessments may provide additional support for individualized treatment decision-making.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4357-4370"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605532/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145514066","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
Fecal microbiota transplantation plus immune checkpoint inhibitor rechallenges in patients with advanced non-small cell lung cancer: a single-arm exploratory study. 粪便微生物群移植加免疫检查点抑制剂在晚期非小细胞肺癌患者中的再挑战:一项单臂探索性研究
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-10-31 Epub Date: 2025-10-27 DOI: 10.21037/tlcr-2025-973
Yijia Du, Yanying Li, Min Yu, Yang Yu, Yan Zhang, Xianfeng Wang, Jianquan He, Lin Lin, Baolong Shen, Youling Gong, Jiang Zhu, Bingwen Zou, Yongmei Liu, Meijuan Huang, You Lu
<p><strong>Background: </strong>The management of advanced non-small cell lung cancer (NSCLC) after progression on initial immunotherapy represents a significant clinical challenge. Immune checkpoint inhibitor (ICI) rechallenge is a considered option, yet its efficacy remains limited. Strategies to enhance the efficacy of ICI rechallenge are urgently needed. There is a pressing need for novel strategies to sensitize tumors to ICI rechallenge. Previous studies have established a correlation between the gut microbiota and the tumor response to immunotherapy, and have explored the application of fecal microbiota transplantation (FMT) in modifying the immune response by restoring the gut microbiota. However, the potential of FMT from healthy donor to reverse immunotherapy resistance in patients with NSCLC has not been previously investigated. This preliminary study aimed to provide initial insights into the safety, tolerability, and potential efficacy of the combined therapy of FMT from healthy donor with immunotherapy rechallenge in NSCLC patients.</p><p><strong>Methods: </strong>In this single-arm exploratory study, patients with advanced NSCLC who progressed after prior immunotherapy were screened and enrolled based on predefined eligibility criteria, including prior response to ICIs and adequate organ function. Eligible patients received oral FMT capsules from healthy donors followed by rechallenge with camrelizumab. The primary endpoint was safety and feasibility [incidence of adverse events (AEs) graded by CTCAE v5.0]. Secondary end points included the objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS). Fecal and blood samples were collected for exploratory analyses of microbiota, metabolome, and T cell receptor (TCR) repertoire.</p><p><strong>Results: </strong>Seven patients (all male, median age 55 years) were enrolled and constituted the analysis population. The majority (5/7) had received three or more prior lines of therapy. During the follow-up period, none experienced grade 4 or higher AEs. The treatment-related AEs were mainly associated with immunotherapy, and only grade 1 FMT-related AEs (e.g., nausea, diarrhea, bloating, and constipation) were reported. One patient achieved a partial response (PR) and one achieved stable disease (SD) with PFS times of 14.6 and 8.1 months, respectively. The median PFS was 1.5 months [95% confidence interval (CI): 1.24-1.75], and the OS was 12.1 months (95% CI: 0.3-23.9) for all patients. Moreover, the treatment modulated the composition of the intestinal flora in all patients, with alpha diversity increasing in responders and decreasing in non-responders.</p><p><strong>Conclusions: </strong>The results indicated that the combined therapy of FMT and immunotherapy rechallenge was feasible and demonstrated a tolerable safety profile in this small cohort. The observed clinical activity is preliminary. These findings support the need for la
背景:在初始免疫治疗进展后的晚期非小细胞肺癌(NSCLC)的管理是一个重大的临床挑战。免疫检查点抑制剂(ICI)再挑战是一种考虑的选择,但其疗效仍然有限。迫切需要提高ICI再挑战有效性的策略。迫切需要新的策略来使肿瘤对ICI再挑战敏感。以往的研究已经建立了肠道微生物群与肿瘤免疫应答之间的相关性,并探索了粪便微生物群移植(FMT)通过恢复肠道微生物群来改变免疫应答的应用。然而,健康供体FMT逆转非小细胞肺癌患者免疫治疗耐药的潜力尚未被研究过。这项初步研究旨在初步了解健康供体FMT与免疫疗法再挑战联合治疗非小细胞肺癌患者的安全性、耐受性和潜在疗效。方法:在这项单臂探索性研究中,根据预先确定的资格标准筛选和入组经过先前免疫治疗后进展的晚期NSCLC患者,包括先前对ICIs的反应和足够的器官功能。符合条件的患者接受健康供体的口服FMT胶囊,然后再接受camrelizumab治疗。主要终点是安全性和可行性[不良事件发生率(ae)按CTCAE v5.0分级]。次要终点包括客观缓解率(ORR)、疾病控制率(DCR)、无进展生存期(PFS)和总生存期(OS)。收集粪便和血液样本,对微生物群、代谢组和T细胞受体(TCR)进行探索性分析。结果:纳入7例患者(均为男性,中位年龄55岁),构成分析人群。大多数患者(5/7)之前接受过三条或更多的治疗。在随访期间,没有人经历4级或更高的ae。治疗相关的不良事件主要与免疫治疗相关,仅报道了1级fmt相关的不良事件(如恶心、腹泻、腹胀和便秘)。1例患者部分缓解(PR), 1例患者病情稳定(SD), PFS时间分别为14.6个月和8.1个月。所有患者的中位PFS为1.5个月[95%可信区间(CI): 1.24-1.75], OS为12.1个月(95% CI: 0.3-23.9)。此外,治疗调节了所有患者肠道菌群的组成,反应者的α多样性增加,无反应者的α多样性减少。结论:结果表明,FMT和免疫治疗再挑战的联合治疗是可行的,并且在这个小队列中显示出可耐受的安全性。观察到的临床活性是初步的。这些发现支持需要更大规模的对照试验来评估这种方法的有效性。
{"title":"Fecal microbiota transplantation plus immune checkpoint inhibitor rechallenges in patients with advanced non-small cell lung cancer: a single-arm exploratory study.","authors":"Yijia Du, Yanying Li, Min Yu, Yang Yu, Yan Zhang, Xianfeng Wang, Jianquan He, Lin Lin, Baolong Shen, Youling Gong, Jiang Zhu, Bingwen Zou, Yongmei Liu, Meijuan Huang, You Lu","doi":"10.21037/tlcr-2025-973","DOIUrl":"10.21037/tlcr-2025-973","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;The management of advanced non-small cell lung cancer (NSCLC) after progression on initial immunotherapy represents a significant clinical challenge. Immune checkpoint inhibitor (ICI) rechallenge is a considered option, yet its efficacy remains limited. Strategies to enhance the efficacy of ICI rechallenge are urgently needed. There is a pressing need for novel strategies to sensitize tumors to ICI rechallenge. Previous studies have established a correlation between the gut microbiota and the tumor response to immunotherapy, and have explored the application of fecal microbiota transplantation (FMT) in modifying the immune response by restoring the gut microbiota. However, the potential of FMT from healthy donor to reverse immunotherapy resistance in patients with NSCLC has not been previously investigated. This preliminary study aimed to provide initial insights into the safety, tolerability, and potential efficacy of the combined therapy of FMT from healthy donor with immunotherapy rechallenge in NSCLC patients.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;In this single-arm exploratory study, patients with advanced NSCLC who progressed after prior immunotherapy were screened and enrolled based on predefined eligibility criteria, including prior response to ICIs and adequate organ function. Eligible patients received oral FMT capsules from healthy donors followed by rechallenge with camrelizumab. The primary endpoint was safety and feasibility [incidence of adverse events (AEs) graded by CTCAE v5.0]. Secondary end points included the objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS). Fecal and blood samples were collected for exploratory analyses of microbiota, metabolome, and T cell receptor (TCR) repertoire.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;Seven patients (all male, median age 55 years) were enrolled and constituted the analysis population. The majority (5/7) had received three or more prior lines of therapy. During the follow-up period, none experienced grade 4 or higher AEs. The treatment-related AEs were mainly associated with immunotherapy, and only grade 1 FMT-related AEs (e.g., nausea, diarrhea, bloating, and constipation) were reported. One patient achieved a partial response (PR) and one achieved stable disease (SD) with PFS times of 14.6 and 8.1 months, respectively. The median PFS was 1.5 months [95% confidence interval (CI): 1.24-1.75], and the OS was 12.1 months (95% CI: 0.3-23.9) for all patients. Moreover, the treatment modulated the composition of the intestinal flora in all patients, with alpha diversity increasing in responders and decreasing in non-responders.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Conclusions: &lt;/strong&gt;The results indicated that the combined therapy of FMT and immunotherapy rechallenge was feasible and demonstrated a tolerable safety profile in this small cohort. The observed clinical activity is preliminary. These findings support the need for la","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4541-4559"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605592/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145514207","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
Doxorubicin paclitaxel in pretreated advanced small-cell lung cancer: a large real-life retrospective study. 阿霉素紫杉醇治疗晚期小细胞肺癌:一项大型现实回顾性研究。
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-10-31 Epub Date: 2025-10-28 DOI: 10.21037/tlcr-2025-653
Coraline Cheuvart, Paul Gougis, Luca Campedel, Clara Bourrachot, Camille Rolland Debord, Baptiste Abbar

Background: Small-cell lung cancer (SCLC) is a highly aggressive disease with limited treatment options beyond first-line platinum-based chemotherapy. Although various second-line regimens such as topotecan and cyclophosphamide-adriamycin-vincristine have been evaluated, clinical outcomes remain poor, particularly in patients with platinum-resistant or refractory disease. The combination of paclitaxel and doxorubicin showed encouraging results in early-phase trials, but data on its use in real-world settings are lacking. This study aimed to assess the efficacy and safety of this chemotherapy regimen in patients with previously treated SCLC in a real-world clinical setting.

Methods: Data were retrospectively collected from all consecutive patients with previously treated SCLC at a single institution in France between 2000 and 2024.

Results: A total of 149 patients were included, of whom 79.9% had platinum-resistant or refractory disease at the time of paclitaxel-doxorubicin initiation. The median progression-free survival (PFS) was 2.4 months [95% confidence interval (CI): 2.1-3.8], and the median overall survival (OS) was 5.1 months (95% CI: 4.7-6.0). The objective response rate was 22.2% in the overall population and 27.7% when excluding patients not evaluable for response. The disease control rate was 41.6% including all patients and 52.1% when excluding non-evaluable cases. In multivariable analysis, an Eastern Cooperative Oncology Group performance status of 2-4 compared to 0-1 was associated with shorter PFS [hazard ratio (HR) =1.9, 95% CI: 1.22-2.8, P=0.004] and OS (HR =2.73, 95% CI: 1.76-4.2, P<0.001). Adverse events led to dose reductions in 54.9% of patients, primarily due to general deterioration, hematologic toxicity, or neuropathy. No treatment-related deaths were reported.

Conclusions: This real-world study suggests that the paclitaxel-doxorubicin combination provides some clinical activity in previously treated SCLC, including in platinum-resistant/refractory disease. Although toxicities were common, they were generally manageable. These findings support further investigation of this regimen with careful patient selection.

背景:小细胞肺癌(SCLC)是一种高度侵袭性疾病,除了一线铂类化疗外,治疗选择有限。虽然各种二线方案,如拓扑替康和环磷酰胺-阿霉素-长春新碱已经被评估过,但临床结果仍然很差,特别是对于铂耐药或难治性疾病的患者。紫杉醇和阿霉素联合使用在早期试验中显示出令人鼓舞的结果,但缺乏在现实环境中使用的数据。本研究旨在评估在现实世界的临床环境中,这种化疗方案对先前治疗过的SCLC患者的有效性和安全性。方法:回顾性收集2000年至2024年在法国一家机构连续接受SCLC治疗的所有患者的数据。结果:共纳入149例患者,其中79.9%的患者在紫杉醇-阿霉素起始治疗时患有铂耐药或难治性疾病。中位无进展生存期(PFS)为2.4个月[95%可信区间(CI): 2.1-3.8],中位总生存期(OS)为5.1个月(95% CI: 4.7-6.0)。在总体人群中,客观缓解率为22.2%,在排除无法评估缓解的患者时,客观缓解率为27.7%。包括所有患者的疾病控制率为41.6%,排除不可评估病例的疾病控制率为52.1%。在多变量分析中,与0-1相比,东部肿瘤合作组的表现状态为2-4与较短的PFS(风险比(HR) =1.9, 95% CI: 1.22-2.8, P=0.004)和OS (HR =2.73, 95% CI: 1.76-4.2)相关。结论:这项现实世界的研究表明,紫杉醇-阿霉素联合治疗对先前治疗过的SCLC,包括铂耐药/难治性疾病有一定的临床活性。虽然毒性很常见,但它们通常是可控的。这些发现支持在仔细选择患者的情况下对该方案进行进一步研究。
{"title":"Doxorubicin paclitaxel in pretreated advanced small-cell lung cancer: a large real-life retrospective study.","authors":"Coraline Cheuvart, Paul Gougis, Luca Campedel, Clara Bourrachot, Camille Rolland Debord, Baptiste Abbar","doi":"10.21037/tlcr-2025-653","DOIUrl":"10.21037/tlcr-2025-653","url":null,"abstract":"<p><strong>Background: </strong>Small-cell lung cancer (SCLC) is a highly aggressive disease with limited treatment options beyond first-line platinum-based chemotherapy. Although various second-line regimens such as topotecan and cyclophosphamide-adriamycin-vincristine have been evaluated, clinical outcomes remain poor, particularly in patients with platinum-resistant or refractory disease. The combination of paclitaxel and doxorubicin showed encouraging results in early-phase trials, but data on its use in real-world settings are lacking. This study aimed to assess the efficacy and safety of this chemotherapy regimen in patients with previously treated SCLC in a real-world clinical setting.</p><p><strong>Methods: </strong>Data were retrospectively collected from all consecutive patients with previously treated SCLC at a single institution in France between 2000 and 2024.</p><p><strong>Results: </strong>A total of 149 patients were included, of whom 79.9% had platinum-resistant or refractory disease at the time of paclitaxel-doxorubicin initiation. The median progression-free survival (PFS) was 2.4 months [95% confidence interval (CI): 2.1-3.8], and the median overall survival (OS) was 5.1 months (95% CI: 4.7-6.0). The objective response rate was 22.2% in the overall population and 27.7% when excluding patients not evaluable for response. The disease control rate was 41.6% including all patients and 52.1% when excluding non-evaluable cases. In multivariable analysis, an Eastern Cooperative Oncology Group performance status of 2-4 compared to 0-1 was associated with shorter PFS [hazard ratio (HR) =1.9, 95% CI: 1.22-2.8, P=0.004] and OS (HR =2.73, 95% CI: 1.76-4.2, P<0.001). Adverse events led to dose reductions in 54.9% of patients, primarily due to general deterioration, hematologic toxicity, or neuropathy. No treatment-related deaths were reported.</p><p><strong>Conclusions: </strong>This real-world study suggests that the paclitaxel-doxorubicin combination provides some clinical activity in previously treated SCLC, including in platinum-resistant/refractory disease. Although toxicities were common, they were generally manageable. These findings support further investigation of this regimen with careful patient selection.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4412-4421"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605157/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145514223","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
Predicting the prognosis of patients with lung adenocarcinoma treated with third-generation EGFR-TKI alone using nomograms based on CT radiomic and clinicopathological factors. 基于CT放射学和临床病理因素的形态图预测单独使用第三代EGFR-TKI治疗肺腺癌患者的预后。
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-10-31 Epub Date: 2025-10-29 DOI: 10.21037/tlcr-2025-763
Junfeng Zhao, Xinhang Gu, Jiaxiao Geng, Ying Li, Yan Wang, Chengxin Liu

Background: Considerable differences exist in the prognosis of patients suffering from advanced non-small cell lung cancer. In fact, they undergo third-generation epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKI) single-agent targeted treatment. This study aimed to recognize the predictive factors influencing the prognosis of such patients, and identify patients at high risk of relapse to instruct clinical therapy and ameliorate prognosis.

Methods: Our research included 255 patients suffering from advanced lung adenocarcinoma (LUAD) treated with third-generation EGFR-TKI monotherapy alone, categorized into the training and validation cohorts. In univariate and multivariate analyses, clinicopathologic features and radiomic features were included. Moreover, statistically significant predictive factors were applied to formulate a nomogram. Area under the curve (AUC) of receiver operating characteristic (ROC) curves, calibration curves, and decision curve analysis (DCA) were utilized to verify the model.

Results: Neutrophil-lymphocyte ratio (NLR) ≥4.6, EGFRex21 L858R mutation, brain metastasis, and radiomic characteristics worked as independent risk factors for progression-free survival (PFS). Age ≥60 years, EGFRex21 L858R mutation, brain metastasis, monocyte-lymphocyte ratio (MLR) ≥0.3, and radiomic features acted as independent risk factors for overall survival (OS). Incorporating this into the nomogram, the AUC of the nomogram for forecasting 6-, 12-, and 24-month PFS and 1-, 2-, and 3-year OS were 0.810, 0.862, 0.873, and 0.886, 0.881, and 0.839, respectively, in the training cohort, and 0.885, 0.858, 0.847 in the validation cohort, and 0.804, 0.824, 0.806, all showing excellent discrimination. The calibration curves showed great consistency between predicted and actual observations. DCA exhibited a gratifying positive net benefit in most threshold probabilities, indicative of a beneficial clinical result. As claimed by PFS and OS survival analysis, the nomogram could distinguish low-risk group from high-risk group.

Conclusions: Our research formulated and corroborated a nomogram, according to clinicopathologic factors and radiomic features. Such a prediction model could be utilized as a strong tool for forecasting the prognosis of patients.

背景:晚期非小细胞肺癌患者的预后存在相当大的差异。事实上,他们接受第三代表皮生长因子受体酪氨酸激酶抑制剂(EGFR-TKI)单药靶向治疗。本研究旨在识别影响该类患者预后的预测因素,识别复发高危患者,指导临床治疗,改善预后。方法:本研究纳入255例接受第三代EGFR-TKI单药治疗的晚期肺腺癌(LUAD)患者,分为训练组和验证组。在单因素和多因素分析中,包括临床病理特征和放射学特征。此外,统计显著的预测因子被应用于形成一个模态图。利用受试者工作特征(ROC)曲线、校准曲线和决策曲线分析(DCA)的曲线下面积(AUC)对模型进行验证。结果:中性粒细胞-淋巴细胞比率(NLR)≥4.6,EGFRex21 L858R突变,脑转移和放射学特征是无进展生存(PFS)的独立危险因素。年龄≥60岁、EGFRex21 L858R突变、脑转移、单核细胞-淋巴细胞比(MLR)≥0.3、放射学特征是总生存(OS)的独立危险因素。将其纳入nomogram中,预测6个月、12个月、24个月PFS和1年、2年、3年OS的nomogram AUC分别为0.810、0.862、0.873和0.886、0.881、0.839,验证队列的AUC分别为0.885、0.858、0.847和0.804、0.824、0.806,均表现出极好的判别性。校正曲线的预测值与实际观测值具有较好的一致性。在大多数阈值概率中,DCA表现出令人满意的正净收益,表明有益的临床结果。根据PFS和OS生存分析,nomogram可以区分低危组和高危组。结论:我们的研究根据临床病理因素和放射学特征制定并证实了一种nomographic。该预测模型可作为预测患者预后的有力工具。
{"title":"Predicting the prognosis of patients with lung adenocarcinoma treated with third-generation EGFR-TKI alone using nomograms based on CT radiomic and clinicopathological factors.","authors":"Junfeng Zhao, Xinhang Gu, Jiaxiao Geng, Ying Li, Yan Wang, Chengxin Liu","doi":"10.21037/tlcr-2025-763","DOIUrl":"10.21037/tlcr-2025-763","url":null,"abstract":"<p><strong>Background: </strong>Considerable differences exist in the prognosis of patients suffering from advanced non-small cell lung cancer. In fact, they undergo third-generation epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKI) single-agent targeted treatment. This study aimed to recognize the predictive factors influencing the prognosis of such patients, and identify patients at high risk of relapse to instruct clinical therapy and ameliorate prognosis.</p><p><strong>Methods: </strong>Our research included 255 patients suffering from advanced lung adenocarcinoma (LUAD) treated with third-generation EGFR-TKI monotherapy alone, categorized into the training and validation cohorts. In univariate and multivariate analyses, clinicopathologic features and radiomic features were included. Moreover, statistically significant predictive factors were applied to formulate a nomogram. Area under the curve (AUC) of receiver operating characteristic (ROC) curves, calibration curves, and decision curve analysis (DCA) were utilized to verify the model.</p><p><strong>Results: </strong>Neutrophil-lymphocyte ratio (NLR) ≥4.6, EGFRex21 L858R mutation, brain metastasis, and radiomic characteristics worked as independent risk factors for progression-free survival (PFS). Age ≥60 years, EGFRex21 L858R mutation, brain metastasis, monocyte-lymphocyte ratio (MLR) ≥0.3, and radiomic features acted as independent risk factors for overall survival (OS). Incorporating this into the nomogram, the AUC of the nomogram for forecasting 6-, 12-, and 24-month PFS and 1-, 2-, and 3-year OS were 0.810, 0.862, 0.873, and 0.886, 0.881, and 0.839, respectively, in the training cohort, and 0.885, 0.858, 0.847 in the validation cohort, and 0.804, 0.824, 0.806, all showing excellent discrimination. The calibration curves showed great consistency between predicted and actual observations. DCA exhibited a gratifying positive net benefit in most threshold probabilities, indicative of a beneficial clinical result. As claimed by PFS and OS survival analysis, the nomogram could distinguish low-risk group from high-risk group.</p><p><strong>Conclusions: </strong>Our research formulated and corroborated a nomogram, according to clinicopathologic factors and radiomic features. Such a prediction model could be utilized as a strong tool for forecasting the prognosis of patients.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4221-4234"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605197/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145513997","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
SuFu somatic mutations are recognized as novel markers for genomic instability and tumor growth in lung adenocarcinoma. SuFu体细胞突变被认为是肺腺癌基因组不稳定性和肿瘤生长的新标记。
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-10-31 Epub Date: 2025-10-29 DOI: 10.21037/tlcr-2025-723
Ji-Hong Liu, Tao Zhang, Yan Wang, Meng-Si Zhang, Jian He, Xiang-Dong Zhou
<p><strong>Background: </strong>Lung adenocarcinoma (LUAD), the predominant histological subtype of non-small cell lung cancer, remains a leading cause of cancer-related mortality worldwide. Suppressor of fused (SuFu) protein, a key negative regulator of the Hedgehog (Hh) signaling pathway, exhibits context-dependent roles in cancer, functioning variably as an oncogene or tumor suppressor. While loss-of-function mutations in SuFu have been implicated in tumorigenesis across several cancer types, their clinical significance in LUAD remains unclear. The study aimed to explore SuFu's regulatory effects in LUAD and evaluate its clinical value as a biomarker.</p><p><strong>Methods: </strong>Endogenous molecular data of SuFu in LUAD were obtained from public bioinformatics databases to investigate SuFu's impact on tumor mutational burden (TMB) and survival. Additionally, immunohistochemical staining was performed to analyze SuFu's impact on clinical features and overall survival (OS). SuFu was knocked out in A549 cells, followed by transcriptome sequencing and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Three mutant (MT) SuFu vectors were constructed. Dual-luciferase assays and Co-immunoprecipitation (Co-IP) experiments were conducted using these vectors to explore the functional interaction between SuFu and Glioma-associated oncogene homolog 1 (Gli1). Overexpressing cell lines of MT SuFu were established in H1975, and the transcription level of Gli1 was detected by real-time quantitative polymerase-chain reaction (RT-qPCR). Overexpressing cell lines of wild type (WT) SuFu were established in A549 and H1975, and the regulatory effect of WT SuFu on the growth of LUAD was verified through functional experiments <i>in vitro</i> and <i>in vivo</i>.</p><p><strong>Results: </strong>The bioinformatics analysis results indicated that SuFu was negatively correlated with TMB in LUAD, while the latter was positively correlated with high-grade node stage (N stage) and higher overall stage. In addition, high expression of SuFu significantly improved the prognosis of LUAD patients, while <i>SuFu</i> mutations significantly reduced the OS. Immunohistochemical results further confirmed that WT SuFu inhibited T stage, N stage, M stage, and overall stage progression, and improved OS. In A549 cells, knocking out SuFu upregulated the response level of the Hh signaling pathway. In LUAD, the endogenous expression of SuFu was not significantly reduced, but it possessed a mutation rate of approximately 1%. The dual-luciferase assays and Co-IP experiments demonstrated that <i>SuFu</i> mutation significantly upregulated the transcriptional activity of Gli1 and downregulated the binding strength with Gli1. RT-qPCR revealed that MT SuFu upregulated Gli1's transcription level. Functional experiments <i>in vitro</i> and <i>in vivo</i> demonstrated the tumor-suppressing effect of WT SuFu.</p><p><strong>Conclusions: </strong>SuFu's rare somatic mutations (~1%)
背景:肺腺癌(LUAD)是非小细胞肺癌的主要组织学亚型,仍然是世界范围内癌症相关死亡的主要原因。融合蛋白抑制因子(SuFu)是Hedgehog (Hh)信号通路的关键负调控因子,在癌症中表现出上下文依赖的作用,可作为癌基因或肿瘤抑制因子发挥不同的作用。虽然SuFu的功能缺失突变与几种癌症类型的肿瘤发生有关,但其在LUAD中的临床意义尚不清楚。本研究旨在探讨素复在LUAD中的调节作用,并评价其作为生物标志物的临床价值。方法:从公共生物信息学数据库中获取素复在LUAD中的内源性分子数据,研究素复对肿瘤突变负荷(TMB)和生存的影响。免疫组化染色分析素复对临床特征和总生存期的影响。在A549细胞中敲除SuFu,然后进行转录组测序和京都基因与基因组百科全书(KEGG)富集分析。构建了三个突变体(MT) SuFu载体。利用这些载体进行双荧光素酶测定和共免疫沉淀(Co-IP)实验,探索素复与胶质瘤相关癌基因同源物1 (Gli1)之间的功能相互作用。1975年建立苏福MT过表达细胞系,采用实时定量聚合酶链反应(RT-qPCR)检测Gli1的转录水平。在A549和H1975中建立野生型(WT)苏福过表达细胞系,通过体外和体内功能实验验证WT苏福对LUAD生长的调节作用。结果:生物信息学分析结果显示,苏复与LUAD患者TMB呈负相关,TMB与高级别淋巴结分期(N期)和高总体分期呈正相关。此外,高表达SuFu可显著改善LUAD患者的预后,而SuFu突变可显著降低OS。免疫组化结果进一步证实,WT素复抑制T期、N期、M期及总分期进展,改善OS。在A549细胞中,敲除SuFu可上调Hh信号通路的应答水平。在LUAD中,SuFu的内源表达没有显著降低,但其突变率约为1%。双荧光素酶分析和Co-IP实验表明,SuFu突变显著上调了Gli1的转录活性,下调了与Gli1的结合强度。RT-qPCR结果显示,苏复可上调Gli1的转录水平。体外和体内功能实验证实了苏复的抑瘤作用。结论:SuFu罕见的体细胞突变(约1%)具有临床价值,可作为LUAD进展的标志,为LUAD的发病机制提供了新的认识。涉及SuFu-Gli1轴的潜在治疗策略也被提出用于精准医学治疗LUAD。
{"title":"<i>SuFu</i> somatic mutations are recognized as novel markers for genomic instability and tumor growth in lung adenocarcinoma.","authors":"Ji-Hong Liu, Tao Zhang, Yan Wang, Meng-Si Zhang, Jian He, Xiang-Dong Zhou","doi":"10.21037/tlcr-2025-723","DOIUrl":"10.21037/tlcr-2025-723","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;Lung adenocarcinoma (LUAD), the predominant histological subtype of non-small cell lung cancer, remains a leading cause of cancer-related mortality worldwide. Suppressor of fused (SuFu) protein, a key negative regulator of the Hedgehog (Hh) signaling pathway, exhibits context-dependent roles in cancer, functioning variably as an oncogene or tumor suppressor. While loss-of-function mutations in SuFu have been implicated in tumorigenesis across several cancer types, their clinical significance in LUAD remains unclear. The study aimed to explore SuFu's regulatory effects in LUAD and evaluate its clinical value as a biomarker.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;Endogenous molecular data of SuFu in LUAD were obtained from public bioinformatics databases to investigate SuFu's impact on tumor mutational burden (TMB) and survival. Additionally, immunohistochemical staining was performed to analyze SuFu's impact on clinical features and overall survival (OS). SuFu was knocked out in A549 cells, followed by transcriptome sequencing and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Three mutant (MT) SuFu vectors were constructed. Dual-luciferase assays and Co-immunoprecipitation (Co-IP) experiments were conducted using these vectors to explore the functional interaction between SuFu and Glioma-associated oncogene homolog 1 (Gli1). Overexpressing cell lines of MT SuFu were established in H1975, and the transcription level of Gli1 was detected by real-time quantitative polymerase-chain reaction (RT-qPCR). Overexpressing cell lines of wild type (WT) SuFu were established in A549 and H1975, and the regulatory effect of WT SuFu on the growth of LUAD was verified through functional experiments &lt;i&gt;in vitro&lt;/i&gt; and &lt;i&gt;in vivo&lt;/i&gt;.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;The bioinformatics analysis results indicated that SuFu was negatively correlated with TMB in LUAD, while the latter was positively correlated with high-grade node stage (N stage) and higher overall stage. In addition, high expression of SuFu significantly improved the prognosis of LUAD patients, while &lt;i&gt;SuFu&lt;/i&gt; mutations significantly reduced the OS. Immunohistochemical results further confirmed that WT SuFu inhibited T stage, N stage, M stage, and overall stage progression, and improved OS. In A549 cells, knocking out SuFu upregulated the response level of the Hh signaling pathway. In LUAD, the endogenous expression of SuFu was not significantly reduced, but it possessed a mutation rate of approximately 1%. The dual-luciferase assays and Co-IP experiments demonstrated that &lt;i&gt;SuFu&lt;/i&gt; mutation significantly upregulated the transcriptional activity of Gli1 and downregulated the binding strength with Gli1. RT-qPCR revealed that MT SuFu upregulated Gli1's transcription level. Functional experiments &lt;i&gt;in vitro&lt;/i&gt; and &lt;i&gt;in vivo&lt;/i&gt; demonstrated the tumor-suppressing effect of WT SuFu.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Conclusions: &lt;/strong&gt;SuFu's rare somatic mutations (~1%)","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4500-4513"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605719/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145514110","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
Deep learning-based classification of pleural malignancy using medical thoracoscopic images. 基于深度学习的胸腔镜胸膜恶性肿瘤分类。
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-10-31 Epub Date: 2025-10-29 DOI: 10.21037/tlcr-2025-588
Yu Jin Hong, Se Hee Ha, Seong Hyeon Park, Jick Hwan Ha, Hyung Woo Kim, Bo Ra Lee, Sang Haak Lee, Chang Dong Yeo, Jong-Yeup Kim, Joon Young Choi

Background: Malignant pleural effusion (MPE) is a frequent complication of advanced lung cancer, and rapid and accurate diagnosis is critical for timely therapeutic decision-making. Although medical thoracoscopy (MT) provides direct visualization and targeted biopsy, resulting in high diagnostic yield, the clinical utility of its findings is contingent on operator experience and subsequent confirmation via pathological analysis. Recent advances in deep learning have enabled automated image classification in various fields, but its application in thoracoscopic images remains unexplored. The aim of our study was to develop a deep learning-based model to classify pleural malignancy and to evaluate its diagnostic performance.

Methods: We developed a deep learning-based classification model using thoracoscopic images obtained from 426 patients who underwent MT at Incheon St. Mary's Hospital between May 2015 and July 2024. After preprocessing and standardization of 4,932 images (2,093 benign, 2,839 malignant), we trained an InceptionV3-based convolutional neural network using transfer learning and online augmentation during training. Model performance was evaluated according to accuracy, precision, recall, the F1 score, the area under the receiver operating characteristic curve (ROC-AUC), and gradient-weighted class activation mapping (Grad-CAM) visualization.

Results: In total, 4,932 thoracoscopic images from 426 patients were used to train and evaluate the model. In the test set, the InceptionV3-based model achieved a classification accuracy of 81.7% [95% confidence interval (CI): ± 3.5%], with a precision of 82.4% (95% CI: ±4.3%), recall of 86.6% (95% CI: ±3.8%), and F1 score of 84.6% (95% CI: ±3.0%). The AUC was 0.90 (95% CI: ±2.6%), indicating excellent discriminative performance. A confusion matrix indicated 165 true positives, 25 false negatives, 29 false positives, and 262 true negatives. Grad-CAM visualizations confirmed that the model consistently focused on visually relevant pleural abnormalities such as nodularity, thickening, and neovascularization. Notably, the model maintained high accuracy even in cases without overt tumor nodules.

Conclusions: This study presents the first deep learning-based classification model of pleural malignancy using MT images. The model showed excellent diagnostic performance and has the potential to aid real-time clinical decision-making during thoracoscopy by suggesting malignant targets for biopsy or pleurodesis.

背景:恶性胸腔积液(MPE)是晚期肺癌的常见并发症,快速准确的诊断对及时做出治疗决策至关重要。虽然医学胸腔镜(MT)提供了直接的可视化和靶向活检,导致高诊断率,但其结果的临床应用取决于操作人员的经验和随后通过病理分析的确认。深度学习的最新进展已经在各个领域实现了自动图像分类,但其在胸腔镜图像中的应用仍未探索。我们研究的目的是开发一个基于深度学习的模型来分类胸膜恶性肿瘤并评估其诊断性能。方法:我们利用2015年5月至2024年7月期间在仁川圣玛丽医院接受MT治疗的426名患者的胸腔镜图像开发了一个基于深度学习的分类模型。在对4932张图像(2093张良性图像,2839张恶性图像)进行预处理和标准化后,我们在训练过程中使用迁移学习和在线增强技术训练了一个基于inception v3的卷积神经网络。根据准确率、精密度、召回率、F1评分、受试者工作特征曲线下面积(ROC-AUC)和梯度加权类激活映射(Grad-CAM)可视化来评估模型的性能。结果:共使用426例患者的4932张胸腔镜图像对模型进行训练和评估。在测试集中,基于inceptionv3的模型分类准确率为81.7%[95%置信区间(CI):±3.5%],准确率为82.4% (95% CI:±4.3%),召回率为86.6% (95% CI:±3.8%),F1评分为84.6% (95% CI:±3.0%)。AUC为0.90 (95% CI:±2.6%),具有良好的鉴别性能。混淆矩阵显示165个真阳性,25个假阴性,29个假阳性,262个真阴性。Grad-CAM可视化证实该模型始终聚焦于视觉相关的胸膜异常,如结节、增厚和新生血管。值得注意的是,即使在没有明显肿瘤结节的情况下,该模型也保持了很高的准确性。结论:本研究提出了首个基于MT图像的胸膜恶性肿瘤深度学习分类模型。该模型表现出优异的诊断性能,并有可能在胸腔镜检查中通过提示活检或胸膜切除术的恶性目标来帮助实时临床决策。
{"title":"Deep learning-based classification of pleural malignancy using medical thoracoscopic images.","authors":"Yu Jin Hong, Se Hee Ha, Seong Hyeon Park, Jick Hwan Ha, Hyung Woo Kim, Bo Ra Lee, Sang Haak Lee, Chang Dong Yeo, Jong-Yeup Kim, Joon Young Choi","doi":"10.21037/tlcr-2025-588","DOIUrl":"10.21037/tlcr-2025-588","url":null,"abstract":"<p><strong>Background: </strong>Malignant pleural effusion (MPE) is a frequent complication of advanced lung cancer, and rapid and accurate diagnosis is critical for timely therapeutic decision-making. Although medical thoracoscopy (MT) provides direct visualization and targeted biopsy, resulting in high diagnostic yield, the clinical utility of its findings is contingent on operator experience and subsequent confirmation via pathological analysis. Recent advances in deep learning have enabled automated image classification in various fields, but its application in thoracoscopic images remains unexplored. The aim of our study was to develop a deep learning-based model to classify pleural malignancy and to evaluate its diagnostic performance.</p><p><strong>Methods: </strong>We developed a deep learning-based classification model using thoracoscopic images obtained from 426 patients who underwent MT at Incheon St. Mary's Hospital between May 2015 and July 2024. After preprocessing and standardization of 4,932 images (2,093 benign, 2,839 malignant), we trained an InceptionV3-based convolutional neural network using transfer learning and online augmentation during training. Model performance was evaluated according to accuracy, precision, recall, the F1 score, the area under the receiver operating characteristic curve (ROC-AUC), and gradient-weighted class activation mapping (Grad-CAM) visualization.</p><p><strong>Results: </strong>In total, 4,932 thoracoscopic images from 426 patients were used to train and evaluate the model. In the test set, the InceptionV3-based model achieved a classification accuracy of 81.7% [95% confidence interval (CI): ± 3.5%], with a precision of 82.4% (95% CI: ±4.3%), recall of 86.6% (95% CI: ±3.8%), and F1 score of 84.6% (95% CI: ±3.0%). The AUC was 0.90 (95% CI: ±2.6%), indicating excellent discriminative performance. A confusion matrix indicated 165 true positives, 25 false negatives, 29 false positives, and 262 true negatives. Grad-CAM visualizations confirmed that the model consistently focused on visually relevant pleural abnormalities such as nodularity, thickening, and neovascularization. Notably, the model maintained high accuracy even in cases without overt tumor nodules.</p><p><strong>Conclusions: </strong>This study presents the first deep learning-based classification model of pleural malignancy using MT images. The model showed excellent diagnostic performance and has the potential to aid real-time clinical decision-making during thoracoscopy by suggesting malignant targets for biopsy or pleurodesis.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4475-4484"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605396/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145514163","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
Diagnostic utility of cryobiopsy for invasive mucinous lung adenocarcinoma. 低温活检对侵袭性肺粘液腺癌的诊断价值。
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-10-31 Epub Date: 2025-10-29 DOI: 10.21037/tlcr-2025-865
Atsushi Kosuge, Yuji Matsumoto, Hideaki Furuse, Kenya Sato, Jumpei Kashima, Satsuki Kishikawa, Masaya Yotsukura, Tomohiro Haruki, Yukihiro Yoshida, Shun-Ichi Watanabe, Yasushi Yatabe, Takaaki Tsuchida

Background: Invasive mucinous lung adenocarcinoma (IMA) is challenging to diagnose via bronchoscopy due to its distinct histopathological characteristics, including poorly atypical tumor cells with abundant mucins. Given that cryobiopsy facilitates high-quality and quantity tissue sampling, we hypothesized that it could improve diagnostic success. This retrospective study aimed to determine whether cryobiopsy is a valid diagnostic tool for IMA.

Methods: We retrospectively reviewed consecutive patients who underwent surgical resection for lung cancer at our institution between March 2017 and July 2024. Among them, patients diagnosed with IMA were selected. Those who had undergone a diagnostic biopsy for peripheral pulmonary lesions (PPLs) were included in the study. These patients were divided into two groups: cryo and conventional, based on whether cryobiopsy was performed. The diagnostic yield and safety profiles of both groups were compared.

Results: Of 5,053 surgeries for lung cancer, 201 (4.0%) were diagnosed with IMA. Among them, 106 patients who had undergone bronchoscopy for PPLs were included in the analyses: 46 in the cryo group and 60 in the conventional group. The diagnostic yield was significantly higher in the cryo group than in the conventional group (93.5% vs. 73.3%, P=0.01). Multivariable analysis showed cryobiopsy was significantly associated with higher yield [adjusted odds ratio (OR), 5.07; 95% confidence interval (CI): 1.33-19.40; P=0.02]. Although not fatal, complications like bleeding and pneumonia were more frequent in the cryo group.

Conclusions: Cryobiopsy is a valid technique for making a diagnosis of IMA.

背景:浸润性肺粘液腺癌(IMA)具有独特的组织病理学特征,包括肿瘤细胞不典型,粘液蛋白丰富,因此通过支气管镜诊断具有挑战性。鉴于低温活检有助于高质量和数量的组织采样,我们假设它可以提高诊断成功率。本回顾性研究旨在确定冷冻活检是否为IMA的有效诊断工具。方法:我们回顾性分析了2017年3月至2024年7月期间在我院接受肺癌手术切除的连续患者。其中选取确诊为IMA的患者。那些接受过外周肺病变诊断活检(ppl)的患者被纳入研究。这些患者根据是否进行冷冻活检分为两组:冷冻组和常规组。比较两组的诊断率和安全性。结果:5053例肺癌手术中,201例(4.0%)被诊断为IMA。其中,106例经支气管镜检查的ppl患者纳入分析:低温组46例,常规组60例。低温组的诊断率明显高于常规组(93.5% vs. 73.3%, P=0.01)。多变量分析显示冷冻活检与高产量显著相关[校正优势比(OR), 5.07;95%置信区间(CI): 1.33-19.40;P = 0.02)。虽然不是致命的,但像出血和肺炎这样的并发症在低温组更常见。结论:冷冻活检是诊断IMA的有效方法。
{"title":"Diagnostic utility of cryobiopsy for invasive mucinous lung adenocarcinoma.","authors":"Atsushi Kosuge, Yuji Matsumoto, Hideaki Furuse, Kenya Sato, Jumpei Kashima, Satsuki Kishikawa, Masaya Yotsukura, Tomohiro Haruki, Yukihiro Yoshida, Shun-Ichi Watanabe, Yasushi Yatabe, Takaaki Tsuchida","doi":"10.21037/tlcr-2025-865","DOIUrl":"10.21037/tlcr-2025-865","url":null,"abstract":"<p><strong>Background: </strong>Invasive mucinous lung adenocarcinoma (IMA) is challenging to diagnose via bronchoscopy due to its distinct histopathological characteristics, including poorly atypical tumor cells with abundant mucins. Given that cryobiopsy facilitates high-quality and quantity tissue sampling, we hypothesized that it could improve diagnostic success. This retrospective study aimed to determine whether cryobiopsy is a valid diagnostic tool for IMA.</p><p><strong>Methods: </strong>We retrospectively reviewed consecutive patients who underwent surgical resection for lung cancer at our institution between March 2017 and July 2024. Among them, patients diagnosed with IMA were selected. Those who had undergone a diagnostic biopsy for peripheral pulmonary lesions (PPLs) were included in the study. These patients were divided into two groups: cryo and conventional, based on whether cryobiopsy was performed. The diagnostic yield and safety profiles of both groups were compared.</p><p><strong>Results: </strong>Of 5,053 surgeries for lung cancer, 201 (4.0%) were diagnosed with IMA. Among them, 106 patients who had undergone bronchoscopy for PPLs were included in the analyses: 46 in the cryo group and 60 in the conventional group. The diagnostic yield was significantly higher in the cryo group than in the conventional group (93.5% <i>vs.</i> 73.3%, P=0.01). Multivariable analysis showed cryobiopsy was significantly associated with higher yield [adjusted odds ratio (OR), 5.07; 95% confidence interval (CI): 1.33-19.40; P=0.02]. Although not fatal, complications like bleeding and pneumonia were more frequent in the cryo group.</p><p><strong>Conclusions: </strong>Cryobiopsy is a valid technique for making a diagnosis of IMA.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4514-4526"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605677/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145514172","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
Deciphering the potential pathogeny of rare tracheal adenoid cystic carcinoma by single-cell RNA-sequencing. 利用单细胞rna测序技术解读罕见气管腺样囊性癌的潜在发病机制。
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-10-31 Epub Date: 2025-10-29 DOI: 10.21037/tlcr-2025-832
Jie Li, Yaping Long, Tao Wang, Haixu Hu, Yanju Yu, Jinfeng Li, Lijuan Zhang, Bo Yang, Yi Liu

Background: Tracheal adenoid cystic carcinoma (TACC) is an extremely rare type of cancer, characterized by slow growth, but high rates of recurrence and metastasis. The treatment of TACC is greatly hampered by the limited understanding of its molecular characteristics. We sought to conduct an in-depth analysis of this type of cancer through single-cell RNA-sequencing (RNA-seq) to provide important evidence for the formulation of treatment strategies for TACC.

Methods: The peripheral blood, tumor tissue, and adjacent tissue of the same TACC patient was obtained and tested by single-cell RNA-seq. A bioinformatics analysis was conducted to identify the cell clones with key biological functions and determine their molecular characteristics. Fluorescent multiplex immunohistochemistry (mIHC) was used to confirm the markers with abnormal expression.

Results: RNA-seq and mIHC verification were successfully performed in one and four TACC patients, respectively. All the sequenced cells were divided into three main categories of epithelial, immune, and stromal cells. In relation to the epithelial cells in the tumor tissue, ciliated cells expressing commonly observed stem-cell markers were found at the beginning of the pseudo-time curve, indicating that they might be related to the origin of the TACC. Further analysis revealed eight genes in the ciliated cells may warrant further investigation. Additionally, the function of immune cells like natural killer (NK) cells and cytotoxic T cells were found to be impaired, which might be the reason for the expansion of TACC. Notably, a distinct subgroup of mesothelial cells disguised as stromal cells was identified. These cells possessed a malignant phenotype and probably interact with macrophages through the MIF-(CD74 + CXCR4 or CD44) pathway. The high density of the CD68+CD74+ cells and their close distance to the Vimentin+PANCK+ cells was confirmed by mIHC. These findings suggest that these markers could be used in the development of drugs targeting TACC.

Conclusions: Our research showed that TACC might originate from ciliated epithelial cells. Abnormal gene expression triggered by external factors, along with the dysfunction of NK and T cells, are critical factors in the genesis and progression of TACC. Developing drugs that target MIF-CD74 signaling pathways may lead to breakthroughs in the treatment of TACC.

背景:气管腺样囊性癌(TACC)是一种极其罕见的肿瘤类型,其特点是生长缓慢,但复发和转移率高。由于对其分子特性的了解有限,对TACC的治疗受到了很大的阻碍。我们试图通过单细胞rna测序(RNA-seq)对这种类型的癌症进行深入分析,为制定TACC的治疗策略提供重要证据。方法:取同一例TACC患者外周血、肿瘤组织及癌旁组织,采用单细胞RNA-seq检测。通过生物信息学分析,鉴定具有关键生物学功能的细胞克隆,确定其分子特征。采用荧光多重免疫组织化学(mIHC)对异常表达的标志物进行确认。结果:分别在1例和4例TACC患者中成功进行了RNA-seq和mIHC验证。所有测序的细胞被分为上皮细胞、免疫细胞和基质细胞三大类。相对于肿瘤组织中的上皮细胞,在伪时间曲线的开始处发现了表达常见干细胞标记物的纤毛细胞,表明它们可能与TACC的起源有关。进一步的分析显示,纤毛细胞中的8个基因可能值得进一步研究。此外,自然杀伤细胞(NK)和细胞毒性T细胞等免疫细胞功能受损,这可能是TACC扩增的原因。值得注意的是,发现了一个伪装成间质细胞的间皮细胞亚群。这些细胞具有恶性表型,可能通过MIF-(CD74 + CXCR4或CD44)途径与巨噬细胞相互作用。mIHC证实CD68+CD74+细胞密度高,与Vimentin+PANCK+细胞距离近。这些发现表明,这些标记物可用于开发靶向TACC的药物。结论:我们的研究表明TACC可能起源于纤毛上皮细胞。外部因素引发的基因表达异常以及NK、T细胞功能障碍是TACC发生发展的关键因素。开发靶向MIF-CD74信号通路的药物可能会导致TACC治疗的突破。
{"title":"Deciphering the potential pathogeny of rare tracheal adenoid cystic carcinoma by single-cell RNA-sequencing.","authors":"Jie Li, Yaping Long, Tao Wang, Haixu Hu, Yanju Yu, Jinfeng Li, Lijuan Zhang, Bo Yang, Yi Liu","doi":"10.21037/tlcr-2025-832","DOIUrl":"10.21037/tlcr-2025-832","url":null,"abstract":"<p><strong>Background: </strong>Tracheal adenoid cystic carcinoma (TACC) is an extremely rare type of cancer, characterized by slow growth, but high rates of recurrence and metastasis. The treatment of TACC is greatly hampered by the limited understanding of its molecular characteristics. We sought to conduct an in-depth analysis of this type of cancer through single-cell RNA-sequencing (RNA-seq) to provide important evidence for the formulation of treatment strategies for TACC.</p><p><strong>Methods: </strong>The peripheral blood, tumor tissue, and adjacent tissue of the same TACC patient was obtained and tested by single-cell RNA-seq. A bioinformatics analysis was conducted to identify the cell clones with key biological functions and determine their molecular characteristics. Fluorescent multiplex immunohistochemistry (mIHC) was used to confirm the markers with abnormal expression.</p><p><strong>Results: </strong>RNA-seq and mIHC verification were successfully performed in one and four TACC patients, respectively. All the sequenced cells were divided into three main categories of epithelial, immune, and stromal cells. In relation to the epithelial cells in the tumor tissue, ciliated cells expressing commonly observed stem-cell markers were found at the beginning of the pseudo-time curve, indicating that they might be related to the origin of the TACC. Further analysis revealed eight genes in the ciliated cells may warrant further investigation. Additionally, the function of immune cells like natural killer (NK) cells and cytotoxic T cells were found to be impaired, which might be the reason for the expansion of TACC. Notably, a distinct subgroup of mesothelial cells disguised as stromal cells was identified. These cells possessed a malignant phenotype and probably interact with macrophages through the MIF-(CD74 + CXCR4 or CD44) pathway. The high density of the CD68<sup>+</sup>CD74<sup>+</sup> cells and their close distance to the Vimentin<sup>+</sup>PANCK<sup>+</sup> cells was confirmed by mIHC. These findings suggest that these markers could be used in the development of drugs targeting TACC.</p><p><strong>Conclusions: </strong>Our research showed that TACC might originate from ciliated epithelial cells. Abnormal gene expression triggered by external factors, along with the dysfunction of NK and T cells, are critical factors in the genesis and progression of TACC. Developing drugs that target MIF-CD74 signaling pathways may lead to breakthroughs in the treatment of TACC.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4460-4474"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605701/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145514180","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
Comparative outcomes of treatment with versus without definitive thoracic radiotherapy in locally advanced inoperable non-small cell lung cancer during the immunotherapy era: a Chinese real-world study. 免疫治疗时代局部晚期不能手术的非小细胞肺癌胸部放疗与不胸部放疗的比较结果:一项中国现实世界研究。
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-10-31 Epub Date: 2025-10-29 DOI: 10.21037/tlcr-2025-701
Xueru Zhu, Jun Liu, Ruxin Cai, Wen Feng, Jun Liu, Wen Yu, Xuwei Cai, Qin Zhang, Xiaolong Fu

Background: The heterogeneity of locally advanced non-small cell lung cancer (LA-NSCLC) has led to varied treatment strategies. Before the immunotherapy era, definitive chemoradiotherapy (CRT) was the standard for inoperable patients. This real-world study evaluates the role of thoracic radiotherapy (RT) in patients receiving first-line immunotherapy-based treatment.

Methods: This retrospective analysis included stage III NSCLC patients from January 2018 to December 2022 who were inoperable or declined surgery and received immunotherapy. Treatment patterns, survival outcomes, and failure modes were assessed. Real-world overall survival (OS), progression-free survival (PFS), and local recurrence-free survival (LRFS) were analyzed using Kaplan-Meier methods.

Results: Among 410 eligible patients, 173 received RT (RT group), while 237 underwent chemoimmunotherapy (CIT) alone (non-RT group). The RT group demonstrated significantly longer median OS (55.5 vs. 26.6 months) and PFS (21.3 vs. 14.1 months) compared to the non-RT group (P<0.001). Patients receiving ≤4 CIT induction cycles before RT had improved outcomes versus >4 cycles (P=0.04). No survival difference was observed between RT doses of 50 Gy and 60 Gy.

Conclusions: This study confirms that thoracic RT remains essential for inoperable LA-NSCLC in the immunotherapy era. An exploratory analysis suggests that a potential strategy could involve ≤4 cycles of CIT induction followed by concurrent CRT (cCRT) and immune checkpoint inhibitor maintenance. These findings encourage further investigation into RT integration within multimodal treatment, with 50-60 Gy appearing potentially comparable in selected patients.

背景:局部晚期非小细胞肺癌(LA-NSCLC)的异质性导致了不同的治疗策略。在免疫治疗时代之前,明确的放化疗(CRT)是不能手术患者的标准。这项真实世界的研究评估了胸部放疗(RT)在接受一线免疫治疗的患者中的作用。方法:回顾性分析2018年1月至2022年12月期间无法手术或拒绝手术并接受免疫治疗的III期NSCLC患者。评估治疗模式、生存结果和失败模式。使用Kaplan-Meier方法分析真实世界总生存期(OS)、无进展生存期(PFS)和局部无复发生存期(LRFS)。结果:在410例符合条件的患者中,173例接受了放疗(放疗组),237例单独接受了化学免疫治疗(CIT)(非放疗组)。与非RT组(P4周期(P=0.04)相比,RT组的中位OS (55.5 vs. 26.6个月)和PFS (21.3 vs. 14.1个月)明显更长。50戈瑞和60戈瑞的放疗剂量之间没有观察到生存差异。结论:本研究证实,在免疫治疗时代,胸部RT对于不能手术的LA-NSCLC仍然是必不可少的。一项探索性分析表明,潜在的策略可能包括≤4个周期的CIT诱导,然后同步CRT (cCRT)和免疫检查点抑制剂维持。这些发现鼓励进一步研究多模式治疗中放疗的整合,在选定的患者中50-60 Gy似乎具有潜在的可比性。
{"title":"Comparative outcomes of treatment with versus without definitive thoracic radiotherapy in locally advanced inoperable non-small cell lung cancer during the immunotherapy era: a Chinese real-world study.","authors":"Xueru Zhu, Jun Liu, Ruxin Cai, Wen Feng, Jun Liu, Wen Yu, Xuwei Cai, Qin Zhang, Xiaolong Fu","doi":"10.21037/tlcr-2025-701","DOIUrl":"10.21037/tlcr-2025-701","url":null,"abstract":"<p><strong>Background: </strong>The heterogeneity of locally advanced non-small cell lung cancer (LA-NSCLC) has led to varied treatment strategies. Before the immunotherapy era, definitive chemoradiotherapy (CRT) was the standard for inoperable patients. This real-world study evaluates the role of thoracic radiotherapy (RT) in patients receiving first-line immunotherapy-based treatment.</p><p><strong>Methods: </strong>This retrospective analysis included stage III NSCLC patients from January 2018 to December 2022 who were inoperable or declined surgery and received immunotherapy. Treatment patterns, survival outcomes, and failure modes were assessed. Real-world overall survival (OS), progression-free survival (PFS), and local recurrence-free survival (LRFS) were analyzed using Kaplan-Meier methods.</p><p><strong>Results: </strong>Among 410 eligible patients, 173 received RT (RT group), while 237 underwent chemoimmunotherapy (CIT) alone (non-RT group). The RT group demonstrated significantly longer median OS (55.5 <i>vs.</i> 26.6 months) and PFS (21.3 <i>vs.</i> 14.1 months) compared to the non-RT group (P<0.001). Patients receiving ≤4 CIT induction cycles before RT had improved outcomes versus >4 cycles (P=0.04). No survival difference was observed between RT doses of 50 Gy and 60 Gy.</p><p><strong>Conclusions: </strong>This study confirms that thoracic RT remains essential for inoperable LA-NSCLC in the immunotherapy era. An exploratory analysis suggests that a potential strategy could involve ≤4 cycles of CIT induction followed by concurrent CRT (cCRT) and immune checkpoint inhibitor maintenance. These findings encourage further investigation into RT integration within multimodal treatment, with 50-60 Gy appearing potentially comparable in selected patients.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4398-4411"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605143/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145514184","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
Evaluation of gefitinib alone or combined with bevacizumab in patients with EGFR L858R-positive advanced non-squamous non-small cell lung cancer: an open-label, randomized, phase 2 trial (BEVA-FLFX-001) with exploratory analysis of plasma biomarkers. 评价吉非替尼单独或联合贝伐单抗治疗EGFR l858r阳性晚期非鳞状非小细胞肺癌:一项开放标签、随机、2期试验(beva - fltex -001),探索性分析血浆生物标志物。
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-10-31 Epub Date: 2025-10-29 DOI: 10.21037/tlcr-2025-545
Xinmin Zhao, Xianghua Wu, Huijie Wang, Hui Yu, Si Sun, Zhihuang Hu, Ying Lin, Yao Zhang, Bo Yu, Zhenhua Wu, Kangli Xiong, Cuicui Liu, Song Wang, Hua Bao, Qiuxiang Ou, Jialei Wang

Background: Patients with advanced non-small cell lung cancer (NSCLC) harboring EGFR L858R mutations have a poorer prognosis than those with exon 19 deletions and exhibit variable responses to first-generation EGFR tyrosine kinase inhibitors (TKIs). Evidence on the use of gefitinib plus bevacizumab in this subgroup remains limited. This study aimed to evaluate the efficacy and safety of gefitinib combined with bevacizumab and to investigate plasma circulating tumor DNA (ctDNA) biomarkers in this patient population.

Methods: In this randomized trial, 81 stage III/IV L858R-mutant NSCLC patients received gefitinib alone or gefitinib plus bevacizumab. The primary endpoint was progression-free survival (PFS), with secondary endpoints including objective response rate (ORR) and safety. Plasma ctDNA was evaluated for prognostic value and detection of treatment-related resistance.

Results: Combination therapy significantly improved median PFS (15.1 vs. 8.2 months, P<0.01) and ORR (90.2% vs. 59.0%, P<0.01) compared to gefitinib alone. Adverse effects were similar, with hypertension occurring exclusively in the combination group. Positive ctDNA at 6 weeks post-treatment correlated with shorter PFS in both groups. A decline in maximal somatic variant allelic frequency (maxVAF) exceeding 98% was associated with improved PFS in the combination group (17.0 vs. 5.4 months, P<0.01). The combination group showed a trend towards lower EGFR T790M mutation frequency (30.8% vs. 58.8%, P=0.16).

Conclusions: Gefitinib plus bevacizumab shows promising efficacy and tolerability in EGFR-mutated NSCLC patients. Plasma ctDNA assessment facilitates the evaluation of treatment efficacy, monitoring disease progression, and informing second-line treatment options.

Trial registration: ClinicalTrials.gov NCT04425187.

背景:携带EGFR L858R突变的晚期非小细胞肺癌(NSCLC)患者的预后比外显子19缺失的患者差,并且对第一代EGFR酪氨酸激酶抑制剂(TKIs)表现出不同的反应。在该亚组中使用吉非替尼加贝伐单抗的证据仍然有限。本研究旨在评估吉非替尼联合贝伐单抗的有效性和安全性,并研究该患者群体的血浆循环肿瘤DNA (ctDNA)生物标志物。方法:在这项随机试验中,81例III/IV期l858r突变的NSCLC患者接受吉非替尼单独或吉非替尼联合贝伐单抗治疗。主要终点是无进展生存期(PFS),次要终点包括客观缓解率(ORR)和安全性。评估血浆ctDNA的预后价值和治疗相关耐药的检测。结果:联合治疗显著提高了中位PFS(15.1个月vs 8.2个月,Pvs. 59.0%, Pvs. 5.4个月),PEGFR T790M突变频率(30.8% vs. 58.8%, P=0.16)。结论:吉非替尼联合贝伐单抗在egfr突变的NSCLC患者中显示出良好的疗效和耐受性。血浆ctDNA评估有助于评估治疗效果,监测疾病进展,并为二线治疗方案提供信息。试验注册:ClinicalTrials.gov NCT04425187。
{"title":"Evaluation of gefitinib alone or combined with bevacizumab in patients with <i>EGFR</i> L858R-positive advanced non-squamous non-small cell lung cancer: an open-label, randomized, phase 2 trial (BEVA-FLFX-001) with exploratory analysis of plasma biomarkers.","authors":"Xinmin Zhao, Xianghua Wu, Huijie Wang, Hui Yu, Si Sun, Zhihuang Hu, Ying Lin, Yao Zhang, Bo Yu, Zhenhua Wu, Kangli Xiong, Cuicui Liu, Song Wang, Hua Bao, Qiuxiang Ou, Jialei Wang","doi":"10.21037/tlcr-2025-545","DOIUrl":"10.21037/tlcr-2025-545","url":null,"abstract":"<p><strong>Background: </strong>Patients with advanced non-small cell lung cancer (NSCLC) harboring <i>EGFR</i> L858R mutations have a poorer prognosis than those with exon 19 deletions and exhibit variable responses to first-generation EGFR tyrosine kinase inhibitors (TKIs). Evidence on the use of gefitinib plus bevacizumab in this subgroup remains limited. This study aimed to evaluate the efficacy and safety of gefitinib combined with bevacizumab and to investigate plasma circulating tumor DNA (ctDNA) biomarkers in this patient population.</p><p><strong>Methods: </strong>In this randomized trial, 81 stage III/IV L858R-mutant NSCLC patients received gefitinib alone or gefitinib plus bevacizumab. The primary endpoint was progression-free survival (PFS), with secondary endpoints including objective response rate (ORR) and safety. Plasma ctDNA was evaluated for prognostic value and detection of treatment-related resistance.</p><p><strong>Results: </strong>Combination therapy significantly improved median PFS (15.1 <i>vs</i>. 8.2 months, P<0.01) and ORR (90.2% <i>vs</i>. 59.0%, P<0.01) compared to gefitinib alone. Adverse effects were similar, with hypertension occurring exclusively in the combination group. Positive ctDNA at 6 weeks post-treatment correlated with shorter PFS in both groups. A decline in maximal somatic variant allelic frequency (maxVAF) exceeding 98% was associated with improved PFS in the combination group (17.0 <i>vs</i>. 5.4 months, P<0.01). The combination group showed a trend towards lower <i>EGFR</i> T790M mutation frequency (30.8% <i>vs</i>. 58.8%, P=0.16).</p><p><strong>Conclusions: </strong>Gefitinib plus bevacizumab shows promising efficacy and tolerability in <i>EGFR</i>-mutated NSCLC patients. Plasma ctDNA assessment facilitates the evaluation of treatment efficacy, monitoring disease progression, and informing second-line treatment options.</p><p><strong>Trial registration: </strong>ClinicalTrials.gov NCT04425187.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4315-4330"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605247/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145514186","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
Translational lung cancer research
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1