首页 > 最新文献

Translational lung cancer research最新文献

英文 中文
Tumor mutation burden and FAT3 mutation influence long-term survival in surgically resected small cell lung cancer. 肿瘤突变负荷和FAT3突变对手术切除小细胞肺癌患者长期生存的影响
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-06-30 Epub Date: 2024-06-27 DOI: 10.21037/tlcr-24-467
Xinyu Qian, Lin Zhu, Na Han, Jing Qin

Background: Small cell lung cancer (SCLC) is highly malignant and has a higher risk of recurrence even in patients who undergo early surgery. However, a subgroup of patients survived for many years. So far, the factors that determine the long-term survivorship remain largely unknown. To determine the genetic characteristics of long-term survival (LTS) after surgery in SCLC, we performed comprehensive comparative genomic profiling and tumor mutation burden (TMB) analysis of resected tumor tissues from patients with LTS and short-term survival (STS) after surgery.

Methods: The present study screened 11 patients from 52 patients with SCLC who underwent surgery at Zhejiang Cancer Hospital from April 2008 to December 2017. A total of six LTS patients (≥4 years) with stage IIB or IIIA SCLC and five STS patients (<2 years) with stage IA or IB SCLC were included in the study. The STS patients were used as a control. All the patients underwent resection without neoadjuvant therapy. We assessed the genomic profiles of the resected tumor tissues and calculated the TMB using next-generation sequencing. We then analyzed and compared the molecular characteristics between the LTS and STS groups.

Results: Our data indicated that tumor tissues from patients with LTS harbor a high TMB. The median TMB for LTS patients was high (approximately 16.4 mutations/Mb), while that for STS patients was low (approximately 8.5 mutations/Mb). The median TMB of patients with LTS and STS showed a trend of significant difference (P=0.08). Gene alterations characterized the survival differences between the two groups. The FAT3 mutation was only found in the LTS group, and the P value determined by Fisher's exact test was 0.06.

Conclusions: A high non-synonymous TMB and the FAT3 mutation could potentially influence LTS after SCLC resection. This study provides valuable information about the molecular differences between LTS and STS patients. Studies with larger sample sizes need to be conducted to confirm our findings in the future.

背景:小细胞肺癌(SCLC)恶性程度高,即使早期接受手术治疗的患者复发风险也较高。然而,也有一部分患者能存活多年。迄今为止,决定长期存活率的因素在很大程度上仍不为人所知。为了确定SCLC术后长期生存(LTS)的基因特征,我们对术后LTS和短期生存(STS)患者的切除肿瘤组织进行了全面的比较基因组图谱分析和肿瘤突变负荷(TMB)分析:本研究从2008年4月至2017年12月在浙江省肿瘤医院接受手术治疗的52例SCLC患者中筛选出11例患者。其中,ⅡB或ⅢA期SCLC患者共6例LTS(≥4岁),STS患者共5例(结果:我们的数据表明,LTS患者的肿瘤组织蕴藏着较高的TMB。LTS患者的中位TMB较高(约16.4个突变/Mb),而STS患者的中位TMB较低(约8.5个突变/Mb)。LTS和STS患者的中位TMB呈显著差异趋势(P=0.08)。基因改变是两组患者生存差异的特征。FAT3基因突变仅在LTS组中发现,费舍尔精确检验的P值为0.06:结论:高非同义TMB和FAT3突变可能会影响SCLC切除术后的LTS。这项研究为 LTS 和 STS 患者的分子差异提供了有价值的信息。未来需要进行样本量更大的研究来证实我们的发现。
{"title":"Tumor mutation burden and <i>FAT3</i> mutation influence long-term survival in surgically resected small cell lung cancer.","authors":"Xinyu Qian, Lin Zhu, Na Han, Jing Qin","doi":"10.21037/tlcr-24-467","DOIUrl":"10.21037/tlcr-24-467","url":null,"abstract":"<p><strong>Background: </strong>Small cell lung cancer (SCLC) is highly malignant and has a higher risk of recurrence even in patients who undergo early surgery. However, a subgroup of patients survived for many years. So far, the factors that determine the long-term survivorship remain largely unknown. To determine the genetic characteristics of long-term survival (LTS) after surgery in SCLC, we performed comprehensive comparative genomic profiling and tumor mutation burden (TMB) analysis of resected tumor tissues from patients with LTS and short-term survival (STS) after surgery.</p><p><strong>Methods: </strong>The present study screened 11 patients from 52 patients with SCLC who underwent surgery at Zhejiang Cancer Hospital from April 2008 to December 2017. A total of six LTS patients (≥4 years) with stage IIB or IIIA SCLC and five STS patients (<2 years) with stage IA or IB SCLC were included in the study. The STS patients were used as a control. All the patients underwent resection without neoadjuvant therapy. We assessed the genomic profiles of the resected tumor tissues and calculated the TMB using next-generation sequencing. We then analyzed and compared the molecular characteristics between the LTS and STS groups.</p><p><strong>Results: </strong>Our data indicated that tumor tissues from patients with LTS harbor a high TMB. The median TMB for LTS patients was high (approximately 16.4 mutations/Mb), while that for STS patients was low (approximately 8.5 mutations/Mb). The median TMB of patients with LTS and STS showed a trend of significant difference (P=0.08). Gene alterations characterized the survival differences between the two groups. The <i>FAT3</i> mutation was only found in the LTS group, and the P value determined by Fisher's exact test was 0.06.</p><p><strong>Conclusions: </strong>A high non-synonymous TMB and the <i>FAT3</i> mutation could potentially influence LTS after SCLC resection. This study provides valuable information about the molecular differences between LTS and STS patients. Studies with larger sample sizes need to be conducted to confirm our findings in the future.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11225038/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141555527","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
Neoadjuvant aumolertinib monotherapy for EGFR-mutant lung squamous cell carcinoma: a case report. 表皮生长因子受体突变肺鳞癌的新辅助奥莫拉替尼单药治疗:病例报告。
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-06-30 Epub Date: 2024-06-20 DOI: 10.21037/tlcr-24-47
Yue Liu, Xiaoxia Yan, Dongliang Bian, Kaixing Ai, Yuming Zhu

Background: Lung cancer is the malignant tumor with high incidence and mortality in China, and more than 30% of non-small cell lung cancer (NSCLC) patients are in the locally advanced stage at the first-time diagnosis. Currently, neoadjuvant epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) combined with radical surgery is effective in the treatment of unresectable stage III EGFR-mutated NSCLC (NSCLCm), and related studies are gradually increasing. But the feasibility of neoadjuvant EGFR-TKI combined with radical surgery for unresectable stage III EGFR-mutant lung squamous cell carcinoma (LUSQm) remains controversial.

Case description: This report presented a successful case of neoadjuvant target-therapy with aumolertinib, the third-generation EGFR-TKI, combined with radical surgery for a stage IIIA LUSQm female patient. After four cycles (28 days/cycle) of neoadjuvant target-therapy, the tumor had a partial response on imaging evaluation and pathological evaluation after surgery showed complete tumor response. The neoadjuvant target-therapy was well tolerated. All adverse events (AEs) that occurred during the treatment were grade I, including decreased platelets, impaired liver function, and diarrhea. The patient was instructed to continue taking Aumolertinib for 3 years after surgery. At the cut-off date of April 1, 2024, the patient had no recurrence after 20 months of treatment.

Conclusions: The result of patient treatment demonstrated the potential feasibility of neoadjuvant Aumolertinib monotherapy for locally advanced LUSQm. The report provides some support for neoadjuvant target-therapy for LUSQm.

背景肺癌是我国发病率和死亡率较高的恶性肿瘤,30%以上的非小细胞肺癌(NSCLC)患者初诊时已处于局部晚期。目前,新辅助表皮生长因子受体酪氨酸激酶抑制剂(EGFR-TKI)联合根治性手术治疗不可切除的Ⅲ期EGFR突变NSCLC(NSCLCm)效果显著,相关研究也逐渐增多。但新辅助EGFR-TKI联合根治性手术治疗不可切除的III期EGFR突变肺鳞状细胞癌(LUSQm)的可行性仍存在争议:本报告介绍了一例第三代表皮生长因子受体抑制剂(EGFR-TKI)奥莫乐替尼(aumolertinib)新辅助靶向治疗联合根治性手术治疗IIIA期LUSQm女性患者的成功案例。经过四个周期(28 天/周期)的新辅助靶向治疗后,肿瘤在影像学评估中出现部分反应,手术后的病理学评估显示肿瘤完全反应。新辅助靶向治疗的耐受性良好。治疗期间发生的所有不良反应(AEs)均为 I 级,包括血小板减少、肝功能受损和腹泻。患者被要求在术后继续服用奥美乐替尼3年。截至2024年4月1日,患者在治疗20个月后没有复发:患者的治疗结果证明了新辅助奥美替尼单药治疗局部晚期LUSQm的潜在可行性。该报告为LUSQm的新辅助靶向治疗提供了一定的支持。
{"title":"Neoadjuvant aumolertinib monotherapy for EGFR-mutant lung squamous cell carcinoma: a case report.","authors":"Yue Liu, Xiaoxia Yan, Dongliang Bian, Kaixing Ai, Yuming Zhu","doi":"10.21037/tlcr-24-47","DOIUrl":"10.21037/tlcr-24-47","url":null,"abstract":"<p><strong>Background: </strong>Lung cancer is the malignant tumor with high incidence and mortality in China, and more than 30% of non-small cell lung cancer (NSCLC) patients are in the locally advanced stage at the first-time diagnosis. Currently, neoadjuvant epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) combined with radical surgery is effective in the treatment of unresectable stage III EGFR-mutated NSCLC (NSCLCm), and related studies are gradually increasing. But the feasibility of neoadjuvant EGFR-TKI combined with radical surgery for unresectable stage III EGFR-mutant lung squamous cell carcinoma (LUSQm) remains controversial.</p><p><strong>Case description: </strong>This report presented a successful case of neoadjuvant target-therapy with aumolertinib, the third-generation EGFR-TKI, combined with radical surgery for a stage IIIA LUSQm female patient. After four cycles (28 days/cycle) of neoadjuvant target-therapy, the tumor had a partial response on imaging evaluation and pathological evaluation after surgery showed complete tumor response. The neoadjuvant target-therapy was well tolerated. All adverse events (AEs) that occurred during the treatment were grade I, including decreased platelets, impaired liver function, and diarrhea. The patient was instructed to continue taking Aumolertinib for 3 years after surgery. At the cut-off date of April 1, 2024, the patient had no recurrence after 20 months of treatment.</p><p><strong>Conclusions: </strong>The result of patient treatment demonstrated the potential feasibility of neoadjuvant Aumolertinib monotherapy for locally advanced LUSQm. The report provides some support for neoadjuvant target-therapy for LUSQm.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11225048/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141555442","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
Correlation analysis between driver gene mutation and clinicopathological features in lung adenocarcinoma based on real-world cumulative clinical data. 基于真实世界累积临床数据的肺腺癌驱动基因突变与临床病理特征相关性分析
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-06-30 Epub Date: 2024-06-27 DOI: 10.21037/tlcr-24-409
Sheng Lu, Aotian Guo, Haichuan Hu, Xinxin Ying, Yao Li, Zhengwei Huang, Wangjue Xu, Shen Tao, Xiaotong Hu, Na Yan, Xuan Zhang, Dan Shen, Takaaki Sasaki, Surein Arulananda, Ken Onodera, Zhengfu He

Background: Driver genes are essential predictors of targeted therapeutic efficacy. Detecting driver gene mutations in lung adenocarcinoma (LUAD) patients can help to screen for targeted drugs and improve patient survival benefits. This study aims to investigate the mutation characterization of driver genes and their correlation with clinicopathological features in LUAD.

Methods: A total of 440 LUAD patients were selected from Sir Run Run Shaw Hospital between July 2019 and September 2022. Postoperative tissue specimens were analyzed for gene mutations using next-generation sequencing technology, focusing, including epidermal growth factor receptor EGFR, ALK, ROS1, RET, KRAS, MET, BRAF, HER2, PIK3CA and NRAS. At the same time, clinicopathological data were collected and organized for multidimensional correlation analysis.

Results: Of 440 LUAD patients, driver gene mutations were not detected in 48 patients. The proportion of patients with driver gene mutations was as high as 89.09%. The top three driver genetic mutations were EGFR, KRAS, and MET. Sixty-nine types of EGFR mutations were detected and distributed in the protein tyrosine kinase catalytic domain (56, 81.16%), Furin-like cysteine-rich region (9, 13.04%), receptor binding domain (3, 4.35%), and EGFR transmembrane domain (1, 1.45%). Single gene locus mutation occurred in 343 LUAD patients, but the mutation gene types covered all tested genes. Our findings showed that EGFR mutations were more commonly observed in non-smoking and female patients (P<0.01), KRAS mutations were more prevalent in male patients and smokers (P<0.01), ROS1 mutations had larger tumor diameters (P<0.01) and RET mutations were more prevalent in smokers (P<0.05).

Conclusions: LUAD patients exhibit diverse genetic mutations, which may co-occur simultaneously. Integrated analysis of multiple mutations is essential for accurate diagnosis and effective treatment of the disease. The use of NGS can significantly expand our understanding of gene mutations and facilitate integrated analysis of multiple gene mutations, providing critical evidence for targeted treatment methods.

背景:驱动基因是靶向治疗疗效的重要预测因子。检测肺腺癌(LUAD)患者的驱动基因突变有助于筛选靶向药物,提高患者生存率。本研究旨在探讨肺腺癌驱动基因的突变特征及其与临床病理特征的相关性:2019年7月至2022年9月期间,邵逸夫医院共选取了440例LUAD患者。采用新一代测序技术对术后组织标本进行基因突变分析,主要包括表皮生长因子受体EGFR、ALK、ROS1、RET、KRAS、MET、BRAF、HER2、PIK3CA和NRAS。同时,还收集整理了临床病理数据,进行多维相关性分析:结果:在440例LUAD患者中,48例未检测到驱动基因突变。有驱动基因突变的患者比例高达 89.09%。前三位驱动基因突变是表皮生长因子受体(EGFR)、KRAS和MET。检测到的69种表皮生长因子受体突变分布在蛋白酪氨酸激酶催化结构域(56种,81.16%)、富含Furin样半胱氨酸区域(9种,13.04%)、受体结合结构域(3种,4.35%)和表皮生长因子受体跨膜结构域(1种,1.45%)。343例LUAD患者发生了单基因位点突变,但突变基因类型涵盖了所有检测基因。我们的研究结果表明,表皮生长因子受体基因突变多见于非吸烟患者和女性患者(PKRAS基因突变多见于男性患者和吸烟者(PRET基因突变多见于吸烟者)):LUAD患者表现出多种基因突变,这些突变可能同时发生。对多种基因突变进行综合分析对于准确诊断和有效治疗疾病至关重要。使用 NGS 可以大大扩展我们对基因突变的了解,促进对多种基因突变的综合分析,为靶向治疗方法提供关键证据。
{"title":"Correlation analysis between driver gene mutation and clinicopathological features in lung adenocarcinoma based on real-world cumulative clinical data.","authors":"Sheng Lu, Aotian Guo, Haichuan Hu, Xinxin Ying, Yao Li, Zhengwei Huang, Wangjue Xu, Shen Tao, Xiaotong Hu, Na Yan, Xuan Zhang, Dan Shen, Takaaki Sasaki, Surein Arulananda, Ken Onodera, Zhengfu He","doi":"10.21037/tlcr-24-409","DOIUrl":"10.21037/tlcr-24-409","url":null,"abstract":"<p><strong>Background: </strong>Driver genes are essential predictors of targeted therapeutic efficacy. Detecting driver gene mutations in lung adenocarcinoma (LUAD) patients can help to screen for targeted drugs and improve patient survival benefits. This study aims to investigate the mutation characterization of driver genes and their correlation with clinicopathological features in LUAD.</p><p><strong>Methods: </strong>A total of 440 LUAD patients were selected from Sir Run Run Shaw Hospital between July 2019 and September 2022. Postoperative tissue specimens were analyzed for gene mutations using next-generation sequencing technology, focusing, including epidermal growth factor receptor <i>EGFR, ALK, ROS1, RET, KRAS, MET, BRAF, HER2, PIK3CA</i> and <i>NRAS</i>. At the same time, clinicopathological data were collected and organized for multidimensional correlation analysis.</p><p><strong>Results: </strong>Of 440 LUAD patients, driver gene mutations were not detected in 48 patients. The proportion of patients with driver gene mutations was as high as 89.09%. The top three driver genetic mutations were <i>EGFR, KRAS</i>, and <i>MET</i>. Sixty-nine types of <i>EGFR</i> mutations were detected and distributed in the protein tyrosine kinase catalytic domain (56, 81.16%), Furin-like cysteine-rich region (9, 13.04%), receptor binding domain (3, 4.35%), and <i>EGFR</i> transmembrane domain (1, 1.45%). Single gene locus mutation occurred in 343 LUAD patients, but the mutation gene types covered all tested genes. Our findings showed that <i>EGFR</i> mutations were more commonly observed in non-smoking and female patients (P<0.01), <i>KRAS</i> mutations were more prevalent in male patients and smokers (P<0.01), ROS1 mutations had larger tumor diameters (P<0.01) and <i>RET</i> mutations were more prevalent in smokers (P<0.05).</p><p><strong>Conclusions: </strong>LUAD patients exhibit diverse genetic mutations, which may co-occur simultaneously. Integrated analysis of multiple mutations is essential for accurate diagnosis and effective treatment of the disease. The use of NGS can significantly expand our understanding of gene mutations and facilitate integrated analysis of multiple gene mutations, providing critical evidence for targeted treatment methods.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11225051/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141555516","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
Diagnosis and treatment of non-small cell lung cancer (NSCLC) harboring MET Ex14 skipping: have we met the desired drug? 跳过 MET Ex14 的非小细胞肺癌 (NSCLC) 的诊断和治疗:我们是否找到了理想的药物?
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-06-30 Epub Date: 2024-06-14 DOI: 10.21037/tlcr-24-93
Go Makimoto
{"title":"Diagnosis and treatment of non-small cell lung cancer (NSCLC) harboring <i>MET</i> Ex14 skipping: have we met the desired drug?","authors":"Go Makimoto","doi":"10.21037/tlcr-24-93","DOIUrl":"10.21037/tlcr-24-93","url":null,"abstract":"","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11225034/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141555517","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 MET exon 14 skipping mutation in pulmonary sarcomatoid carcinoma by whole-tumour texture analysis combined with clinical and conventional contrast-enhanced computed tomography features. 通过全肿瘤纹理分析结合临床和常规对比增强计算机断层扫描特征预测肺肉瘤样癌中的MET 14外显子跳变。
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-06-30 Epub Date: 2024-06-11 DOI: 10.21037/tlcr-24-56
Lei Miao, Tian Qiu, Yan Li, Jianwei Li, Xu Jiang, Mengwen Liu, Xue Zhang, Jiuming Jiang, Huanhuan Zhang, Yanmei Wang, Xiao Li, Jianming Ying, Meng Li

Background: Pulmonary sarcomatoid carcinoma (PSC) is a rare, highly malignant type of non-small cell lung cancer (NSCLC) with a poor prognosis. Targeted drugs for MET exon 14 (METex14) skipping mutation can have considerable clinical benefits. This study aimed to predict METex14 skipping mutation in PSC patients by whole-tumour texture analysis combined with clinical and conventional contrast-enhanced computed tomography (CECT) features.

Methods: This retrospective study included 56 patients with PSC diagnosed by pathology. All patients underwent CECT before surgery or other treatment, and both targeted DNA- and RNA-based next-generation sequencing (NGS) were used to detect METex14 skipping mutation status. The patients were divided into two groups: METex14 skipping mutation and nonmutation groups. Overall, 1,316 texture features of the whole tumour were extracted. We also collected 12 clinical and 20 conventional CECT features. After dimensionality reduction and selection, predictive models were established by multivariate logistic regression analysis. Models were evaluated using the area under the curve (AUC), and the clinical utility of the model was assessed by decision curve analysis.

Results: METex14 skipping mutation was detected in 17.9% of PSCs. Mutations were found more frequently in those (I) who had smaller long- or short-axis diameters (P=0.02, P=0.01); (II) who had lower T stages (I, II) (P=0.02); and (III) with pseudocapsular or annular enhancement (P=0.03). The combined model based on the conventional and texture models yielded the best performance in predicting METex14 skipping mutation with the highest AUC (0.89). The conventional and texture models also had good performance (AUC =0.83 conventional; =0.88 texture).

Conclusions: Whole-tumour texture analysis combined with clinical and conventional CECT features may serve as a noninvasive tool to predict the METex14 skipping mutation status in PSC.

背景:肺肉瘤样癌(PSC)是一种罕见、高度恶性的非小细胞肺癌(NSCLC),预后较差。针对MET外显子14(METex14)跳过突变的靶向药物可产生可观的临床疗效。本研究旨在通过全肿瘤纹理分析,结合临床和常规对比增强计算机断层扫描(CECT)特征,预测PSC患者的METex14跳越突变:这项回顾性研究纳入了56例经病理诊断的PSC患者。所有患者在手术或其他治疗前均接受了CECT检查,并采用基于DNA和RNA的下一代测序(NGS)检测METex14跳变突变状态。患者被分为两组:METex14 跳越突变组和非突变组。总共提取了 1316 个肿瘤纹理特征。我们还收集了 12 个临床特征和 20 个常规 CECT 特征。经过降维和筛选,我们通过多变量逻辑回归分析建立了预测模型。使用曲线下面积(AUC)对模型进行评估,并通过决策曲线分析评估模型的临床实用性:结果:在17.9%的PSCs中检测到了METex14跳越突变。在以下情况中更常发现突变:(I)长轴或短轴直径较小(P=0.02,P=0.01);(II)T分期较低(I,II)(P=0.02);(III)假囊性或环状强化(P=0.03)。基于传统模型和纹理模型的组合模型在预测 METex14 跳变方面表现最佳,AUC 最高(0.89)。传统模型和纹理模型也有很好的表现(传统模型的AUC=0.83;纹理模型的AUC=0.88):结论:全瘤纹理分析与临床和常规CECT特征相结合,可作为预测PSC患者METex14跳跃突变状态的无创工具。
{"title":"Predicting <i>MET</i> exon 14 skipping mutation in pulmonary sarcomatoid carcinoma by whole-tumour texture analysis combined with clinical and conventional contrast-enhanced computed tomography features.","authors":"Lei Miao, Tian Qiu, Yan Li, Jianwei Li, Xu Jiang, Mengwen Liu, Xue Zhang, Jiuming Jiang, Huanhuan Zhang, Yanmei Wang, Xiao Li, Jianming Ying, Meng Li","doi":"10.21037/tlcr-24-56","DOIUrl":"10.21037/tlcr-24-56","url":null,"abstract":"<p><strong>Background: </strong>Pulmonary sarcomatoid carcinoma (PSC) is a rare, highly malignant type of non-small cell lung cancer (NSCLC) with a poor prognosis. Targeted drugs for <i>MET</i> exon 14 (<i>MET</i>ex14) skipping mutation can have considerable clinical benefits. This study aimed to predict <i>MET</i>ex14 skipping mutation in PSC patients by whole-tumour texture analysis combined with clinical and conventional contrast-enhanced computed tomography (CECT) features.</p><p><strong>Methods: </strong>This retrospective study included 56 patients with PSC diagnosed by pathology. All patients underwent CECT before surgery or other treatment, and both targeted DNA- and RNA-based next-generation sequencing (NGS) were used to detect <i>MET</i>ex14 skipping mutation status. The patients were divided into two groups: <i>MET</i>ex14 skipping mutation and nonmutation groups. Overall, 1,316 texture features of the whole tumour were extracted. We also collected 12 clinical and 20 conventional CECT features. After dimensionality reduction and selection, predictive models were established by multivariate logistic regression analysis. Models were evaluated using the area under the curve (AUC), and the clinical utility of the model was assessed by decision curve analysis.</p><p><strong>Results: </strong><i>MET</i>ex14 skipping mutation was detected in 17.9% of PSCs. Mutations were found more frequently in those (I) who had smaller long- or short-axis diameters (P=0.02, P=0.01); (II) who had lower T stages (I, II) (P=0.02); and (III) with pseudocapsular or annular enhancement (P=0.03). The combined model based on the conventional and texture models yielded the best performance in predicting <i>MET</i>ex14 skipping mutation with the highest AUC (0.89). The conventional and texture models also had good performance (AUC =0.83 conventional; =0.88 texture).</p><p><strong>Conclusions: </strong>Whole-tumour texture analysis combined with clinical and conventional CECT features may serve as a noninvasive tool to predict the <i>MET</i>ex14 skipping mutation status in PSC.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11225047/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141555445","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
Predictive value of delta-radiomic features for prognosis of advanced non-small cell lung cancer patients undergoing immune checkpoint inhibitor therapy. 接受免疫检查点抑制剂治疗的晚期非小细胞肺癌患者预后的δ-放射特征预测价值。
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-06-30 Epub Date: 2024-06-12 DOI: 10.21037/tlcr-24-7
Xiaoyu Han, Yujin Wang, Xi Jia, Yuting Zheng, Chengyu Ding, Xiaohui Zhang, Kailu Zhang, Yunkun Cao, Yumin Li, Liming Xia, Chuansheng Zheng, Jing Huang, Heshui Shi

Background: No robust predictive biomarkers exist to identify non-small cell lung cancer (NSCLC) patients likely to benefit from immune checkpoint inhibitor (ICI) therapies. The aim of this study was to explore the role of delta-radiomics features in predicting the clinical outcomes of patients with advanced NSCLC who received ICI therapy.

Methods: Data of 179 patients with advanced NSCLC (stages IIIB-IV) from two institutions (Database 1 =133; Database 2 =46) were retrospectively analyzed. Patients in the Database 1 were randomly assigned into training and validation dataset, with a ratio of 8:2. Patients in Database 2 were allocated into testing dataset. Features were selected from computed tomography (CT) images before and 6-8 weeks after ICI therapy. For each lesion, a total of 1,037 radiomic features were extracted. Lowly reliable [intraclass correlation coefficient (ICC) <0.8] and redundant (r>0.8) features were excluded. The delta-radiomics features were defined as the relative net change of radiomics features between two time points. Prognostic models for progression-free survival (PFS) and overall survival (OS) were established using the multivariate Cox regression based on selected delta-radiomics features. A clinical model and a pre-treatment radiomics model were established as well.

Results: The median PFS (after therapy) was 7.0 [interquartile range (IQR): 3.4, 9.1] (range, 1.4-13.2) months. To predict PFS, the model established based on the five most contributing delta-radiomics features yielded Harrell's concordance index (C-index) values of 0.708, 0.688, and 0.603 in the training, validation, and testing databases, respectively. The median survival time was 12 (IQR: 8.7, 15.8) (range, 2.9-23.3) months. To predict OS, a promising prognostic performance was confirmed with the corresponding C-index values of 0.810, 0.762, and 0.697 in the three datasets based on the seven most contributing delta-radiomics features, respectively. Furthermore, compared with clinical and pre-treatment radiomics models, the delta-radiomics model had the highest area under the curve (AUC) value and the best patients' stratification ability.

Conclusions: The delta-radiomics model showed a good performance in predicting therapeutic outcomes in advanced NSCLC patients undergoing ICI therapy. It provides a higher predictive value than clinical and the pre-treatment radiomics models.

背景:目前尚无可靠的预测性生物标志物来识别可能从免疫检查点抑制剂(ICI)疗法中获益的非小细胞肺癌(NSCLC)患者。本研究旨在探索δ-放射组学特征在预测接受ICI治疗的晚期NSCLC患者临床结局中的作用:方法:对两家机构(数据库1 =133;数据库2 =46)的179名晚期NSCLC(IIIB-IV期)患者的数据进行回顾性分析。数据库1中的患者被随机分配到训练和验证数据集,比例为8:2。数据库 2 中的患者被分配到测试数据集。特征选自 ICI 治疗前和治疗后 6-8 周的计算机断层扫描(CT)图像。每个病灶共提取了 1,037 个放射学特征。可靠性低(类内相关系数(ICC)0.8)的特征被排除在外。delta放射组学特征被定义为两个时间点之间放射组学特征的相对净变化。根据选定的δ-放射组学特征,采用多变量考克斯回归法建立了无进展生存期(PFS)和总生存期(OS)的预后模型。同时还建立了临床模型和治疗前放射组学模型:中位 PFS(治疗后)为 7.0 个月[四分位距(IQR):3.4,9.1](范围:1.4-13.2)。为了预测患者的生存期,根据五个最有贡献的δ-放射组学特征建立的模型在训练、验证和测试数据库中的哈雷尔一致性指数(C-index)值分别为 0.708、0.688 和 0.603。中位生存时间为12个月(IQR:8.7,15.8)(范围:2.9-23.3)。在预测OS方面,基于七个最有贡献的delta放射组学特征,三个数据集的相应C指数值分别为0.810、0.762和0.697,证实了其良好的预后性能。此外,与临床和治疗前放射组学模型相比,delta-放射组学模型的曲线下面积(AUC)值最高,患者分层能力最强:结论:δ-放射组学模型在预测接受ICI治疗的晚期NSCLC患者的治疗结果方面表现良好。它比临床模型和治疗前放射组学模型具有更高的预测价值。
{"title":"Predictive value of delta-radiomic features for prognosis of advanced non-small cell lung cancer patients undergoing immune checkpoint inhibitor therapy.","authors":"Xiaoyu Han, Yujin Wang, Xi Jia, Yuting Zheng, Chengyu Ding, Xiaohui Zhang, Kailu Zhang, Yunkun Cao, Yumin Li, Liming Xia, Chuansheng Zheng, Jing Huang, Heshui Shi","doi":"10.21037/tlcr-24-7","DOIUrl":"10.21037/tlcr-24-7","url":null,"abstract":"<p><strong>Background: </strong>No robust predictive biomarkers exist to identify non-small cell lung cancer (NSCLC) patients likely to benefit from immune checkpoint inhibitor (ICI) therapies. The aim of this study was to explore the role of delta-radiomics features in predicting the clinical outcomes of patients with advanced NSCLC who received ICI therapy.</p><p><strong>Methods: </strong>Data of 179 patients with advanced NSCLC (stages IIIB-IV) from two institutions (Database 1 =133; Database 2 =46) were retrospectively analyzed. Patients in the Database 1 were randomly assigned into training and validation dataset, with a ratio of 8:2. Patients in Database 2 were allocated into testing dataset. Features were selected from computed tomography (CT) images before and 6-8 weeks after ICI therapy. For each lesion, a total of 1,037 radiomic features were extracted. Lowly reliable [intraclass correlation coefficient (ICC) <0.8] and redundant (r>0.8) features were excluded. The delta-radiomics features were defined as the relative net change of radiomics features between two time points. Prognostic models for progression-free survival (PFS) and overall survival (OS) were established using the multivariate Cox regression based on selected delta-radiomics features. A clinical model and a pre-treatment radiomics model were established as well.</p><p><strong>Results: </strong>The median PFS (after therapy) was 7.0 [interquartile range (IQR): 3.4, 9.1] (range, 1.4-13.2) months. To predict PFS, the model established based on the five most contributing delta-radiomics features yielded Harrell's concordance index (C-index) values of 0.708, 0.688, and 0.603 in the training, validation, and testing databases, respectively. The median survival time was 12 (IQR: 8.7, 15.8) (range, 2.9-23.3) months. To predict OS, a promising prognostic performance was confirmed with the corresponding C-index values of 0.810, 0.762, and 0.697 in the three datasets based on the seven most contributing delta-radiomics features, respectively. Furthermore, compared with clinical and pre-treatment radiomics models, the delta-radiomics model had the highest area under the curve (AUC) value and the best patients' stratification ability.</p><p><strong>Conclusions: </strong>The delta-radiomics model showed a good performance in predicting therapeutic outcomes in advanced NSCLC patients undergoing ICI therapy. It provides a higher predictive value than clinical and the pre-treatment radiomics models.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11225045/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141555447","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
The combination of osimertinib and savolitinib as molecular inhibition of EGFR and MET receptors may be selected to provide maximum effectiveness and acceptable toxicity. 可选择奥希替尼和沙沃利替尼联合作为表皮生长因子受体和 MET 受体的分子抑制剂,以提供最大疗效和可接受的毒性。
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-06-30 Epub Date: 2024-06-25 DOI: 10.21037/tlcr-24-204
Dariusz M Kowalski, Magdalena Zaborowska-Szmit, Sebastian Szmit, Piotr Jaśkiewicz, Maciej Krzakowski
{"title":"The combination of osimertinib and savolitinib as molecular inhibition of EGFR and MET receptors may be selected to provide maximum effectiveness and acceptable toxicity.","authors":"Dariusz M Kowalski, Magdalena Zaborowska-Szmit, Sebastian Szmit, Piotr Jaśkiewicz, Maciej Krzakowski","doi":"10.21037/tlcr-24-204","DOIUrl":"10.21037/tlcr-24-204","url":null,"abstract":"","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11225044/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141555524","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
Peptidyl-prolyl isomerase F as a prognostic biomarker associated with immune infiltrates and mitophagy in lung adenocarcinoma. 肽基脯氨酰异构酶F是与肺腺癌免疫浸润和有丝分裂相关的预后生物标志物
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-06-30 Epub Date: 2024-06-20 DOI: 10.21037/tlcr-24-344
Zitong Feng, Lin Yuan, Luyuan Ma, Wenhao Yu, Fayez Kheir, Lukas Käsmann, Wolfgang M Brueckl, Kai Jin, Dingxin Wang, Yi Shen, Rongyang Li, Hui Tian

Background: Lung adenocarcinoma (LUAD) is among the most prevalent malignancies worldwide, with unfavorable treatment outcomes. Peptidyl-prolyl isomerase F (PPIF) is known to influence the malignancy traits of tumor progression by modulating the bioenergetics and mitochondrial permeability in cancer cells; however, its role in LUAD remains unclear. Our study seeks to investigate the clinical significance, tumor proliferation, and immune regulatory functions of PPIF in LUAD.

Methods: The expression of PPIF in LUAD tissues and cells was assessed using bioinformatics analysis, immunohistochemistry (IHC), and Western blotting. Survival curve analysis was conducted to examine the prognostic association between PPIF expression and LUAD. The immunomodulatory role of PPIF in LUAD was assessed through the analysis of PPIF expression and immune cell infiltration. A series of gain- and loss-of-function experiments were conducted on PPIF to investigate its biological functions in LUAD both in vitro and in vivo. The mechanisms underlying PPIF's effects on LUAD were delineated through functional enrichment analysis and Western blotting assays.

Results: PPIF exhibited overexpression in LUAD tissues compared to normal controls. Survival curve analysis revealed that patients with LUAD exhibiting higher PPIF expression demonstrated decreased overall survival and a shorter progression-free interval. PPIF was implicated in modulating immune cell infiltration, particularly in regulating the T helper 1-T helper 2 cell balance. Functionally, PPIF was discovered to promote tumor cell proliferation and advance cell-cycle progression. Furthermore, PPIF could impede mitophagy by targeting the FOXO3a/PINK1-Parkin signaling pathway.

Conclusions: The findings of this study indicate that the prognosis-related gene PPIF may have a significant role in the regulation of LUAD cell proliferation, tumor-associated immune cell infiltration, and mitophagy, and thus PPIF may be a promising therapeutic target of LUAD.

背景:肺腺癌(LUAD)是全球发病率最高的恶性肿瘤之一,治疗效果不佳。众所周知,肽基脯氨酰异构酶 F(PPIF)可通过调节癌细胞的生物能和线粒体通透性来影响肿瘤进展的恶性特征;然而,它在肺腺癌中的作用仍不清楚。我们的研究旨在探讨 PPIF 在 LUAD 中的临床意义、肿瘤增殖和免疫调节功能:方法:采用生物信息学分析、免疫组织化学(IHC)和 Western 印迹法评估 PPIF 在 LUAD 组织和细胞中的表达。研究人员对PPIF的表达与LUAD的预后关系进行了生存曲线分析。通过分析 PPIF 表达和免疫细胞浸润,评估了 PPIF 在 LUAD 中的免疫调节作用。对PPIF进行了一系列功能增益和功能缺失实验,以研究其在体外和体内LUAD中的生物学功能。通过功能富集分析和Western印迹分析,研究了PPIF对LUAD的影响机制:结果:与正常对照组相比,PPIF在LUAD组织中呈现过表达。生存曲线分析表明,PPIF表达较高的LUAD患者总生存率下降,无进展间隔时间缩短。PPIF 参与调节免疫细胞浸润,尤其是调节 T 辅助细胞 1-T 辅助细胞 2 的平衡。在功能上,研究发现 PPIF 能促进肿瘤细胞增殖并推进细胞周期的进展。此外,PPIF还能通过靶向FOXO3a/PINK1-Parkin信号通路阻碍有丝分裂:本研究结果表明,预后相关基因PPIF可能在LUAD细胞增殖、肿瘤相关免疫细胞浸润和有丝分裂的调控中发挥重要作用,因此PPIF可能是LUAD的一个有前景的治疗靶点。
{"title":"Peptidyl-prolyl isomerase F as a prognostic biomarker associated with immune infiltrates and mitophagy in lung adenocarcinoma.","authors":"Zitong Feng, Lin Yuan, Luyuan Ma, Wenhao Yu, Fayez Kheir, Lukas Käsmann, Wolfgang M Brueckl, Kai Jin, Dingxin Wang, Yi Shen, Rongyang Li, Hui Tian","doi":"10.21037/tlcr-24-344","DOIUrl":"10.21037/tlcr-24-344","url":null,"abstract":"<p><strong>Background: </strong>Lung adenocarcinoma (LUAD) is among the most prevalent malignancies worldwide, with unfavorable treatment outcomes. Peptidyl-prolyl isomerase F (<i>PPIF</i>) is known to influence the malignancy traits of tumor progression by modulating the bioenergetics and mitochondrial permeability in cancer cells; however, its role in LUAD remains unclear. Our study seeks to investigate the clinical significance, tumor proliferation, and immune regulatory functions of <i>PPIF</i> in LUAD.</p><p><strong>Methods: </strong>The expression of <i>PPIF</i> in LUAD tissues and cells was assessed using bioinformatics analysis, immunohistochemistry (IHC), and Western blotting. Survival curve analysis was conducted to examine the prognostic association between <i>PPIF</i> expression and LUAD. The immunomodulatory role of <i>PPIF</i> in LUAD was assessed through the analysis of <i>PPIF</i> expression and immune cell infiltration. A series of gain- and loss-of-function experiments were conducted on <i>PPIF</i> to investigate its biological functions in LUAD both <i>in vitro</i> and <i>in vivo</i>. The mechanisms underlying <i>PPIF</i>'s effects on LUAD were delineated through functional enrichment analysis and Western blotting assays.</p><p><strong>Results: </strong><i>PPIF</i> exhibited overexpression in LUAD tissues compared to normal controls. Survival curve analysis revealed that patients with LUAD exhibiting higher <i>PPIF</i> expression demonstrated decreased overall survival and a shorter progression-free interval. <i>PPIF</i> was implicated in modulating immune cell infiltration, particularly in regulating the T helper 1-T helper 2 cell balance. Functionally, <i>PPIF</i> was discovered to promote tumor cell proliferation and advance cell-cycle progression. Furthermore, <i>PPIF</i> could impede mitophagy by targeting the FOXO3a/PINK1-Parkin signaling pathway.</p><p><strong>Conclusions: </strong>The findings of this study indicate that the prognosis-related gene <i>PPIF</i> may have a significant role in the regulation of LUAD cell proliferation, tumor-associated immune cell infiltration, and mitophagy, and thus <i>PPIF</i> may be a promising therapeutic target of LUAD.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11225036/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141555444","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
Bioinformatics analysis of an immunotherapy responsiveness-related gene signature in predicting lung adenocarcinoma prognosis. 预测肺腺癌预后的免疫疗法反应性相关基因特征的生物信息学分析
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-06-30 Epub Date: 2024-06-07 DOI: 10.21037/tlcr-24-309
Yupeng Jiang, Bacha Hammad, Hong Huang, Chenzi Zhang, Bing Xiao, Linxia Liu, Qimi Liu, Hengxing Liang, Zhenyu Zhao, Yawen Gao

Background: Immune therapy has become first-line treatment option for patients with lung cancer, but some patients respond poorly to immune therapy, especially among patients with lung adenocarcinoma (LUAD). Novel tools are needed to screen potential responders to immune therapy in LUAD patients, to better predict the prognosis and guide clinical decision-making. Although many efforts have been made to predict the responsiveness of LUAD patients, the results were limited. During the era of immunotherapy, this study attempts to construct a novel prognostic model for LUAD by utilizing differentially expressed genes (DEGs) among patients with differential immune therapy responses.

Methods: Transcriptome data of 598 patients with LUAD were downloaded from The Cancer Genome Atlas (TCGA) database, which included 539 tumor samples and 59 normal control samples, with a mean follow-up time of 29.69 months (63.1% of patients remained alive by the end of follow-up). Other data sources including three datasets from the Gene Expression Omnibus (GEO) database were analyzed, and the DEGs between immunotherapy responders and nonresponders were identified and screened. Univariate Cox regression analysis was applied with the TCGA cohort as the training set and GSE72094 cohort as the validation set, and least absolute shrinkage and selection operator (LASSO) Cox regression were applied in the prognostic-related genes which fulfilled the filter criteria to establish a prognostic formula, which was then tested with time-dependent receiver operating characteristic (ROC) analysis. Enriched pathways of the prognostic-related genes were analyzed with Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses, and tumor immune microenvironment (TIME), tumor mutational burden, and drug sensitivity tests were completed with appropriate packages in R (The R Foundation of Statistical Computing). Finally, a nomogram incorporating the prognostic formula was established.

Results: A total of 1,636 DEGs were identified, 1,163 prognostic-related DEGs were extracted, and 34 DEGs were selected and incorporated into the immunotherapy responsiveness-related risk score (IRRS) formula. The IRRS formula had good performance in predicting the overall prognoses in patients with LUAD and had excellent performance in prognosis prediction in all LUAD subgroups. Moreover, the IRRS formula could predict anticancer drug sensitivity and immunotherapy responsiveness in patients with LUAD. Mechanistically, immune microenvironments varied profoundly between the two IRRS groups; the most significantly varied pathway between the high-IRRS and low-IRRS groups was ribonucleoprotein complex biogenesis, which correlated closely with the TP53 and TTN mutation burdens. In addition, we established a nomogram incorporating the IRRS, age, sex, clinical stage, T-stage, N-stage, and M-stage as predictors

背景:免疫疗法已成为肺癌患者的一线治疗选择,但有些患者对免疫疗法反应不佳,尤其是肺腺癌(LUAD)患者。我们需要新的工具来筛选肺腺癌患者对免疫疗法的潜在反应者,以便更好地预测预后并指导临床决策。尽管人们已经做了很多努力来预测 LUAD 患者的反应性,但结果都很有限。在免疫疗法大行其道的时代,本研究试图利用免疫疗法反应不同的患者中的差异表达基因(DEGs)构建一个新的LUAD预后模型:从癌症基因组图谱(TCGA)数据库下载了598例LUAD患者的转录组数据,其中包括539个肿瘤样本和59个正常对照样本,平均随访时间为29.69个月(63.1%的患者在随访结束时仍然存活)。研究人员还分析了其他数据源,包括基因表达总库(GEO)数据库中的三个数据集,并确定和筛选了免疫治疗应答者和非应答者之间的 DEGs。以TCGA队列为训练集、GSE72094队列为验证集进行单变量Cox回归分析,并对符合筛选条件的预后相关基因进行最小绝对收缩和选择算子(LASSO)Cox回归,建立预后公式,然后用时间依赖性接收者操作特征(ROC)分析进行检验。预后相关基因的富集通路通过基因本体(GO)和京都基因组百科全书(KEGG)富集分析进行分析,肿瘤免疫微环境(TIME)、肿瘤突变负荷和药物敏感性测试则通过R(The R Foundation of Statistical Computing)中的相应软件包完成。最后,建立了一个包含预后公式的提名图:结果:共鉴定出1,636个DEGs,提取出1,163个与预后相关的DEGs,并筛选出34个DEGs纳入免疫治疗反应性相关风险评分(IRRS)公式。IRRS公式在预测LUAD患者的总体预后方面表现良好,在预测所有LUAD亚组的预后方面表现优异。此外,IRRS公式还能预测LUAD患者的抗癌药物敏感性和免疫治疗反应性。从机理上讲,两个IRRS组之间的免疫微环境差异很大;高IRRS组和低IRRS组之间差异最大的途径是核糖核蛋白复合物的生物生成,这与TP53和TTN突变负荷密切相关。此外,我们还建立了一个将IRRS、年龄、性别、临床分期、T期、N期和M期作为预测因子的提名图,该提名图可以预测LUAD患者1年、3年和5年的预后,其曲线下面积(AUC)分别为0.718、0.702和0.68:本研究建立的模型可以预测LUAD患者的预后,有助于识别对抗癌药物和免疫疗法反应良好的患者,是指导临床决策的重要工具。
{"title":"Bioinformatics analysis of an immunotherapy responsiveness-related gene signature in predicting lung adenocarcinoma prognosis.","authors":"Yupeng Jiang, Bacha Hammad, Hong Huang, Chenzi Zhang, Bing Xiao, Linxia Liu, Qimi Liu, Hengxing Liang, Zhenyu Zhao, Yawen Gao","doi":"10.21037/tlcr-24-309","DOIUrl":"10.21037/tlcr-24-309","url":null,"abstract":"<p><strong>Background: </strong>Immune therapy has become first-line treatment option for patients with lung cancer, but some patients respond poorly to immune therapy, especially among patients with lung adenocarcinoma (LUAD). Novel tools are needed to screen potential responders to immune therapy in LUAD patients, to better predict the prognosis and guide clinical decision-making. Although many efforts have been made to predict the responsiveness of LUAD patients, the results were limited. During the era of immunotherapy, this study attempts to construct a novel prognostic model for LUAD by utilizing differentially expressed genes (DEGs) among patients with differential immune therapy responses.</p><p><strong>Methods: </strong>Transcriptome data of 598 patients with LUAD were downloaded from The Cancer Genome Atlas (TCGA) database, which included 539 tumor samples and 59 normal control samples, with a mean follow-up time of 29.69 months (63.1% of patients remained alive by the end of follow-up). Other data sources including three datasets from the Gene Expression Omnibus (GEO) database were analyzed, and the DEGs between immunotherapy responders and nonresponders were identified and screened. Univariate Cox regression analysis was applied with the TCGA cohort as the training set and GSE72094 cohort as the validation set, and least absolute shrinkage and selection operator (LASSO) Cox regression were applied in the prognostic-related genes which fulfilled the filter criteria to establish a prognostic formula, which was then tested with time-dependent receiver operating characteristic (ROC) analysis. Enriched pathways of the prognostic-related genes were analyzed with Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses, and tumor immune microenvironment (TIME), tumor mutational burden, and drug sensitivity tests were completed with appropriate packages in R (The R Foundation of Statistical Computing). Finally, a nomogram incorporating the prognostic formula was established.</p><p><strong>Results: </strong>A total of 1,636 DEGs were identified, 1,163 prognostic-related DEGs were extracted, and 34 DEGs were selected and incorporated into the immunotherapy responsiveness-related risk score (IRRS) formula. The IRRS formula had good performance in predicting the overall prognoses in patients with LUAD and had excellent performance in prognosis prediction in all LUAD subgroups. Moreover, the IRRS formula could predict anticancer drug sensitivity and immunotherapy responsiveness in patients with LUAD. Mechanistically, immune microenvironments varied profoundly between the two IRRS groups; the most significantly varied pathway between the high-IRRS and low-IRRS groups was ribonucleoprotein complex biogenesis, which correlated closely with the <i>TP53</i> and <i>TTN</i> mutation burdens. In addition, we established a nomogram incorporating the IRRS, age, sex, clinical stage, T-stage, N-stage, and M-stage as predictors","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11225057/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141555514","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
Radical resection in a patient with stage IIIA non-small cell lung cancer with the EGFR exon 19 deletion mutation after neoadjuvant targeted therapy with osimertinib: a case report. 一名表皮生长因子受体外显子19缺失突变的IIIA期非小细胞肺癌患者在使用奥希替尼进行新辅助靶向治疗后接受根治性切除术:病例报告。
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-06-30 Epub Date: 2024-06-27 DOI: 10.21037/tlcr-24-403
Hao Chen, Jiarong Zhang, Atsushi Osoegawa, Lorenzo Calvetti, Palma Fedele, Chun Chen, Bin Zheng

Background: With the advent of targeted therapies, the survival rates of patients with locally advanced lung cancer have significantly improved. However, there is limited research on the efficacy of neoadjuvant targeted therapy in resectable advanced non-small cell lung cancer (NSCLC) patients with positive driver genes. This article reports a case of stage IIIA NSCLC with an epidermal growth factor receptor (EGFR) 19del mutation that successfully underwent radical lung cancer surgery following neoadjuvant targeted therapy. By observing the perioperative treatment outcomes and side effects in this patient, we aimed to provide insights and summarize experiences for treating similar cases in the future.

Case description: We report a case of a 54-year-old female diagnosed preoperatively with stage IIIA adenocarcinoma of the left upper lung (cT1cN2M0). The patient's course was complicated by acute sick sinus syndrome and was cured by implanting a permanent pacemaker. After multidisciplinary discussion, it was decided to administer neoadjuvant targeted therapy with osimertinib. Following 6 weeks of treatment, the tumor assessment showed partial response (PR), making the patient eligible for surgery. The patient underwent single-port thoracoscopic left upper lobectomy + mediastinal lymphadenectomy. Intraoperatively, the left hilar lymph nodes were found to be tightly adherent to the apical-anterior branch of the left upper pulmonary artery. The main trunk of the left pulmonary artery was temporarily occluded with a vascular clamp to safely dissect the left upper pulmonary artery. The procedure was completed without conversion to open thoracotomy, achieving an R0 resection. Postoperative pathology confirmed stage IIIA (ypT1bN2M0), and the patient continued adjuvant therapy with osimertinib.

Conclusions: Neoadjuvant targeted therapy with osimertinib is expected to become one of the options for neoadjuvant therapy in locally advanced NSCLC with sensitizing EGFR mutations. And for those with advanced lung cancer involving tumors close to the hilum or mediastinal lymph node metastasis, preblocking of the left upper pulmonary artery can help improve surgical safety and better ensure R0 resection.

背景:随着靶向疗法的出现,局部晚期肺癌患者的生存率已显著提高。然而,对于驱动基因阳性的可切除晚期非小细胞肺癌(NSCLC)患者,新辅助靶向治疗的疗效研究有限。本文报道了一例表皮生长因子受体(EGFR)19del突变的IIIA期NSCLC患者在接受新辅助靶向治疗后成功接受了肺癌根治手术。通过观察该患者围手术期的治疗效果和副作用,我们旨在为今后治疗类似病例提供见解和总结经验:我们报告了一例术前诊断为左上肺腺癌 IIIA 期(cT1cN2M0)的 54 岁女性患者。患者的病程因急性病窦综合征而变得复杂,通过植入永久起搏器才得以治愈。经过多学科讨论,决定使用奥希替尼进行新辅助靶向治疗。治疗6周后,肿瘤评估显示出现部分反应(PR),因此患者符合手术条件。患者接受了单孔胸腔镜左上叶切除术+纵隔淋巴结切除术。术中发现,左侧肺门淋巴结与左上肺动脉尖前支紧密粘连。用血管钳暂时闭塞了左肺动脉主干,以便安全地解剖左上肺动脉。手术在未转为开胸手术的情况下完成,实现了 R0 切除。术后病理证实为IIIA期(ypT1bN2M0),患者继续接受奥希替尼的辅助治疗:结论:奥希莫替尼新辅助靶向治疗有望成为表皮生长因子受体突变局部晚期NSCLC新辅助治疗的选择之一。对于肿瘤靠近肺门或纵隔淋巴结转移的晚期肺癌患者,预先阻断左上肺动脉有助于提高手术安全性,更好地确保R0切除。
{"title":"Radical resection in a patient with stage IIIA non-small cell lung cancer with the <i>EGFR</i> exon 19 deletion mutation after neoadjuvant targeted therapy with osimertinib: a case report.","authors":"Hao Chen, Jiarong Zhang, Atsushi Osoegawa, Lorenzo Calvetti, Palma Fedele, Chun Chen, Bin Zheng","doi":"10.21037/tlcr-24-403","DOIUrl":"10.21037/tlcr-24-403","url":null,"abstract":"<p><strong>Background: </strong>With the advent of targeted therapies, the survival rates of patients with locally advanced lung cancer have significantly improved. However, there is limited research on the efficacy of neoadjuvant targeted therapy in resectable advanced non-small cell lung cancer (NSCLC) patients with positive driver genes. This article reports a case of stage IIIA NSCLC with an epidermal growth factor receptor (<i>EGFR</i>) 19del mutation that successfully underwent radical lung cancer surgery following neoadjuvant targeted therapy. By observing the perioperative treatment outcomes and side effects in this patient, we aimed to provide insights and summarize experiences for treating similar cases in the future.</p><p><strong>Case description: </strong>We report a case of a 54-year-old female diagnosed preoperatively with stage IIIA adenocarcinoma of the left upper lung (cT1cN2M0). The patient's course was complicated by acute sick sinus syndrome and was cured by implanting a permanent pacemaker. After multidisciplinary discussion, it was decided to administer neoadjuvant targeted therapy with osimertinib. Following 6 weeks of treatment, the tumor assessment showed partial response (PR), making the patient eligible for surgery. The patient underwent single-port thoracoscopic left upper lobectomy + mediastinal lymphadenectomy. Intraoperatively, the left hilar lymph nodes were found to be tightly adherent to the apical-anterior branch of the left upper pulmonary artery. The main trunk of the left pulmonary artery was temporarily occluded with a vascular clamp to safely dissect the left upper pulmonary artery. The procedure was completed without conversion to open thoracotomy, achieving an R0 resection. Postoperative pathology confirmed stage IIIA (ypT1bN2M0), and the patient continued adjuvant therapy with osimertinib.</p><p><strong>Conclusions: </strong>Neoadjuvant targeted therapy with osimertinib is expected to become one of the options for neoadjuvant therapy in locally advanced NSCLC with sensitizing <i>EGFR</i> mutations. And for those with advanced lung cancer involving tumors close to the hilum or mediastinal lymph node metastasis, preblocking of the left upper pulmonary artery can help improve surgical safety and better ensure R0 resection.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11225049/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141555521","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: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