首页 > 最新文献

Translational lung cancer research最新文献

英文 中文
Comparative effectiveness and safety of systemic therapies for treatment-naïve, PD-L1 expression <1% advanced NSCLC: a systematic review and network meta-analysis. 对于treatment-naïve, PD-L1表达<1%的晚期NSCLC,全身治疗的比较有效性和安全性:一项系统综述和网络荟萃分析
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-11-30 Epub Date: 2025-11-27 DOI: 10.21037/tlcr-2025-371
Mengyun Zhou, Junfang Huang, Zhou Jin, Qingqing Hao, Xueying Li, Kunyao Yu, Kunyan Sun, Xiang Zhao, Meng Zhang, Guangfa Wang, Yuan Cheng

Background: For advanced non-small cell lung cancer (NSCLC) with programmed cell death ligand 1 (PD-L1) expression <1% and no actionable oncogenic alterations, pembrolizumab plus chemotherapy (Pembro-chemo) is widely regarded as the current standard of care. However, emerging therapeutic combinations and preliminary results from ongoing trials challenge its superiority, particularly across different histologic types. Therefore, this study aimed to appraise the effectiveness and safety of first-line treatment for PD-L1 <1% advanced, non-squamous and squamous NSCLC.

Methods: PubMed, Ovid Medline, the Cochrane Library, and Embase were searched from database inception to August 15, 2025, to identify phase III randomized controlled trials (RCTs) that explored first-line treatments in treatment-naïve advanced NSCLC, PD-L1 <1%, no epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) alterations; and reported any efficacy outcome were eligible for inclusion. Treatment effectiveness was quantified using overall survival (OS), progression-free survival (PFS) and objective response rate (ORR). Surface under the cumulative ranking value (SUCRA) was used to rank the therapies. Risk of bias for included RCTs was assessed using the Cochrane Risk of Bias 2 tool.

Results: Twenty-five phase III RCTs involving 5,815 participants were eligible. Overall, 21 first-line treatments were identified. In terms of OS, pembrolizumab + chemotherapy + canakinumab (Pembro-chemo-canakinumab) (SUCRA =0.90) showed great potential in improving outcomes, although its long-term efficacy still needed to be validated. Nivolumab + ipilimumab (Nivo-ipi) (SUCRA =0.78) closely followed. Both top regimens showed non-significant superiority over Pembro-chemo. Regarding PFS, nivolumab + chemotherapy + bevacizumab (SUCRA =0.88), and serplulimab + chemotherapy (SUCRA =0.87) were the optimal regimens. Specifically for non-squamous patients, Pembro-chemo was optimal for OS (SUCRA =0.90), followed by Nivolumab + chemotherapy + bevacizumab (SUCRA =0.82). Nivolumab + chemotherapy + bevacizumab optimized PFS, with an hazard ratio (HR) of 0.52 [95% confidence interval (CI): 0.30-0.92 vs. Pembro-chemo]. For squamous patients, nivolumab + ipilimumab ± chemotherapy (Nivo-ipi-chemo) led in OS, while serplulimab + chemotherapy in PFS.

Conclusions: First-line personalized treatment for PD-L1 <1%, advanced NSCLC should be histology-based, balancing efficacy and toxicity. Pembro-chemo and nivolumab + chemotherapy + bevacizumab combinations are recommended as the optimal first-line options for non-squamous patients, and Nivo-ipi-chemo for squamous patients.

背景:对于程序性细胞死亡配体1 (PD-L1)表达的晚期非小细胞肺癌(NSCLC)方法:检索PubMed, Ovid Medline, Cochrane Library和Embase数据库,从数据库建立到2025年8月15日,以确定探索treatment-naïve晚期NSCLC, PD-L1一线治疗的III期随机对照试验(rct)结果:25个III期rct涉及5,815名参与者符合条件。总的来说,确定了21种一线治疗方法。在OS方面,pembrolizumab +化疗+ canakinumab (pembrolizumab -chemo-canakinumab) (SUCRA =0.90)在改善预后方面显示出巨大的潜力,尽管其长期疗效仍有待验证。Nivolumab + ipilimumab (Nivo-ipi) (SUCRA =0.78)紧随其后。两种方案均未显示出与pembrom -chemo相比的显著优势。在PFS方面,纳武单抗+化疗+贝伐单抗(SUCRA =0.88)和塞普利单抗+化疗(SUCRA =0.87)是最优方案。特别是对于非鳞状病变患者,pembrom - chemotherapy是OS的最佳方案(SUCRA =0.90),其次是Nivolumab + chemotherapy + bevacizumab (SUCRA =0.82)。纳武单抗+化疗+贝伐单抗优化PFS,风险比(HR)为0.52[95%置信区间(CI): 0.30-0.92 vs. pembrom -chemo]。对于鳞状患者,nivolumab + ipilimumab±化疗(Nivo-ipi-chemo)在OS中领先,而serpluliumab +化疗在PFS中领先。结论:PD-L1的一线个性化治疗
{"title":"Comparative effectiveness and safety of systemic therapies for treatment-naïve, PD-L1 expression <1% advanced NSCLC: a systematic review and network meta-analysis.","authors":"Mengyun Zhou, Junfang Huang, Zhou Jin, Qingqing Hao, Xueying Li, Kunyao Yu, Kunyan Sun, Xiang Zhao, Meng Zhang, Guangfa Wang, Yuan Cheng","doi":"10.21037/tlcr-2025-371","DOIUrl":"10.21037/tlcr-2025-371","url":null,"abstract":"<p><strong>Background: </strong>For advanced non-small cell lung cancer (NSCLC) with programmed cell death ligand 1 (PD-L1) expression <1% and no actionable oncogenic alterations, pembrolizumab plus chemotherapy (Pembro-chemo) is widely regarded as the current standard of care. However, emerging therapeutic combinations and preliminary results from ongoing trials challenge its superiority, particularly across different histologic types. Therefore, this study aimed to appraise the effectiveness and safety of first-line treatment for PD-L1 <1% advanced, non-squamous and squamous NSCLC.</p><p><strong>Methods: </strong>PubMed, Ovid Medline, the Cochrane Library, and Embase were searched from database inception to August 15, 2025, to identify phase III randomized controlled trials (RCTs) that explored first-line treatments in treatment-naïve advanced NSCLC, PD-L1 <1%, no epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) alterations; and reported any efficacy outcome were eligible for inclusion. Treatment effectiveness was quantified using overall survival (OS), progression-free survival (PFS) and objective response rate (ORR). Surface under the cumulative ranking value (SUCRA) was used to rank the therapies. Risk of bias for included RCTs was assessed using the Cochrane Risk of Bias 2 tool.</p><p><strong>Results: </strong>Twenty-five phase III RCTs involving 5,815 participants were eligible. Overall, 21 first-line treatments were identified. In terms of OS, pembrolizumab + chemotherapy + canakinumab (Pembro-chemo-canakinumab) (SUCRA =0.90) showed great potential in improving outcomes, although its long-term efficacy still needed to be validated. Nivolumab + ipilimumab (Nivo-ipi) (SUCRA =0.78) closely followed. Both top regimens showed non-significant superiority over Pembro-chemo. Regarding PFS, nivolumab + chemotherapy + bevacizumab (SUCRA =0.88), and serplulimab + chemotherapy (SUCRA =0.87) were the optimal regimens. Specifically for non-squamous patients, Pembro-chemo was optimal for OS (SUCRA =0.90), followed by Nivolumab + chemotherapy + bevacizumab (SUCRA =0.82). Nivolumab + chemotherapy + bevacizumab optimized PFS, with an hazard ratio (HR) of 0.52 [95% confidence interval (CI): 0.30-0.92 <i>vs.</i> Pembro-chemo]. For squamous patients, nivolumab + ipilimumab ± chemotherapy (Nivo-ipi-chemo) led in OS, while serplulimab + chemotherapy in PFS.</p><p><strong>Conclusions: </strong>First-line personalized treatment for PD-L1 <1%, advanced NSCLC should be histology-based, balancing efficacy and toxicity. Pembro-chemo and nivolumab + chemotherapy + bevacizumab combinations are recommended as the optimal first-line options for non-squamous patients, and Nivo-ipi-chemo for squamous patients.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"4849-4867"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683440/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715832","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
PD-L1 expression in primary lung tumor as a superior predictive biomarker to metastatic lymph nodes for first-line immunochemotherapy in advanced KRAS-mutant non-small cell lung cancer. 原发性肺肿瘤中PD-L1表达作为晚期kras突变型非小细胞肺癌一线免疫化疗转移淋巴结的优越预测生物标志物
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-11-30 Epub Date: 2025-11-26 DOI: 10.21037/tlcr-2025-817
Hongping Jin, Yidan Zhang, Honglei Huang, Jianlin Xu, Yingqi Xu, Yiqing Wu, Tengfei Liu, Hua Zhong, Jie Chen, Qian Miao, Runbo Zhong

Background: The correlation between programmed death-ligand 1 (PD-L1) expression and the efficacy of first-line chemoimmunotherapy in advanced KRAS-mutant non-small cell lung cancer (NSCLC) is inconsistent across studies, possibly due to the spatial heterogeneity of PD-L1 expression. We characterized the PD-L1 expression profile at different tumor sites and its impact on the clinical efficacy of first-line immunochemotherapy in advanced KRAS-mutant NSCLC patients.

Methods: A retrospective analysis was performed on 302 patients with advanced KRAS-mutant NSCLC who received first-line immunochemotherapy from January 2018 to December 2022. Patients were categorized by biopsy sites: primary lung lesions (n=211) and metastatic lymph nodes (n=91). PD-L1 expression was stratified into <1%, 1-49%, and ≥50%. Primary endpoints included progression-free survival (PFS), objective response rate (ORR), and disease control rate (DCR).

Results: Median PFS was 9.07 months overall, 7.27 months for PD-L1 <1%, 8.30 months for PD-L1 =1-49%, and 15.00 months for PD-L1 ≥50% (P<0.001). The corresponding median PFS values for patients with primary lung lesions were 7.23, 8.30, and 15.03 months, which were statistically significant (P=0.001). In contrast, no significant differences in PFS were observed among the PD-L1 subgroups for patients with metastatic lymph nodes (P=0.17). Moreover, the non-G12C subgroup exhibited a more pronounced PD-L1 expression heterogeneity between the primary and metastatic sites than the G12C subgroup.

Conclusions: In advanced KRAS-mutant NSCLC, the PD-L1 expression in primary lung lesions may predict first-line immunochemotherapy efficacy, while PD-L1 expression in metastatic lymph nodes lacks predictive value, especially in non-G12C mutations.

背景:程序性死亡配体1 (programmed death-ligand 1, PD-L1)表达与晚期kras突变型非小细胞肺癌(NSCLC)一线化疗免疫治疗疗效之间的相关性在各研究中并不一致,可能是由于PD-L1表达的空间异质性。我们分析了不同肿瘤部位PD-L1表达谱及其对晚期kras突变型NSCLC患者一线免疫化疗临床疗效的影响。方法:回顾性分析2018年1月至2022年12月接受一线免疫化疗的302例晚期kras突变NSCLC患者。患者按活检部位分类:原发性肺病变(n=211)和转移性淋巴结(n=91)。结果:中位PFS为9.07个月,PD-L1为7.27个月。结论:在晚期kras突变型NSCLC中,原发性肺病变中PD-L1表达可预测一线免疫化疗疗效,而转移性淋巴结中PD-L1表达缺乏预测价值,特别是在非g12c突变中。
{"title":"PD-L1 expression in primary lung tumor as a superior predictive biomarker to metastatic lymph nodes for first-line immunochemotherapy in advanced KRAS-mutant non-small cell lung cancer.","authors":"Hongping Jin, Yidan Zhang, Honglei Huang, Jianlin Xu, Yingqi Xu, Yiqing Wu, Tengfei Liu, Hua Zhong, Jie Chen, Qian Miao, Runbo Zhong","doi":"10.21037/tlcr-2025-817","DOIUrl":"10.21037/tlcr-2025-817","url":null,"abstract":"<p><strong>Background: </strong>The correlation between programmed death-ligand 1 (PD-L1) expression and the efficacy of first-line chemoimmunotherapy in advanced KRAS-mutant non-small cell lung cancer (NSCLC) is inconsistent across studies, possibly due to the spatial heterogeneity of PD-L1 expression. We characterized the PD-L1 expression profile at different tumor sites and its impact on the clinical efficacy of first-line immunochemotherapy in advanced KRAS-mutant NSCLC patients.</p><p><strong>Methods: </strong>A retrospective analysis was performed on 302 patients with advanced KRAS-mutant NSCLC who received first-line immunochemotherapy from January 2018 to December 2022. Patients were categorized by biopsy sites: primary lung lesions (n=211) and metastatic lymph nodes (n=91). PD-L1 expression was stratified into <1%, 1-49%, and ≥50%. Primary endpoints included progression-free survival (PFS), objective response rate (ORR), and disease control rate (DCR).</p><p><strong>Results: </strong>Median PFS was 9.07 months overall, 7.27 months for PD-L1 <1%, 8.30 months for PD-L1 =1-49%, and 15.00 months for PD-L1 ≥50% (P<0.001). The corresponding median PFS values for patients with primary lung lesions were 7.23, 8.30, and 15.03 months, which were statistically significant (P=0.001). In contrast, no significant differences in PFS were observed among the PD-L1 subgroups for patients with metastatic lymph nodes (P=0.17). Moreover, the non-G12C subgroup exhibited a more pronounced PD-L1 expression heterogeneity between the primary and metastatic sites than the G12C subgroup.</p><p><strong>Conclusions: </strong>In advanced KRAS-mutant NSCLC, the PD-L1 expression in primary lung lesions may predict first-line immunochemotherapy efficacy, while PD-L1 expression in metastatic lymph nodes lacks predictive value, especially in non-G12C mutations.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"4923-4941"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683442/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715857","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
Subtype-specific RNA sequencing using micro-dissection revealed extracellular matrix alterations as key factors in lung adenocarcinoma invasion. 利用显微解剖技术进行的亚型特异性RNA测序揭示了细胞外基质改变是肺腺癌侵袭的关键因素。
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-11-30 Epub Date: 2025-11-27 DOI: 10.21037/tlcr-2025-520
Chongze Yuan, Xiao Chen, Yizhou Peng, Qiang Zheng, Yunjian Pan, Xuxia Shen, Xiaoting Tao, Xingxin Yao, Hui Hong, Hongbin Ji, Yawei Zhang, Yihua Sun

Background: Tumor invasion is a critical step in tumorigenesis and represents an important therapeutic target. However, the molecular mechanisms underlying the invasion process of lung adenocarcinoma (LUAD) remain poorly understood. In this study, we investigated the transcriptomic and epigenetic alterations occurring during LUAD invasion to elucidate the key biological processes.

Methods: Frozen section of LUAD tumors, which contained both invasive and non-invasive subtypes, was selected as the study model. These subtypes were identified, micro-dissected, and separately processed for RNA sequencing (RNA-seq). Subsequent analysis identified differentially expressed genes (DEGs) and significantly enriched biological processes. Additionally, RNA-seq data from independent LUAD tissues were analyzed to screen for critical genes, and H3K27ac chromatin immunoprecipitation sequencing (ChIP-seq) was conducted to explore potential regulatory mechanisms. Finally, survival analysis was performed using The Cancer Genome Atlas (TCGA) database to validate the clinical relevance of the identified genes.

Results: Subtype-specific RNA-seq analysis revealed that alteration in the extracellular matrix (ECM) was a key hallmark of LUAD invasion, which was validated by transcriptomic changes in independent tissue samples. Furthermore, several of these ECM genes were significantly associated with LUAD prognosis. Based on these findings, we hypothesized that epigenetic alterations during LUAD progression may drive these ECM changes, and that subsequent remodeling of the immune microenvironment may also contribute to the invasive process.

Conclusions: Our integrated analysis demonstrated that epigenetic and transcriptomic dysregulation-induced ECM alterations were critical for LUAD invasion. Specifically, key ECM genes, including AGER and CGNL1, were identified as central regulators of invasion and thus represent promising therapeutic targets for LUAD.

背景:肿瘤侵袭是肿瘤发生的关键步骤,是肿瘤治疗的重要靶点。然而,肺腺癌(LUAD)侵袭过程的分子机制仍然知之甚少。在这项研究中,我们研究了LUAD侵袭过程中发生的转录组学和表观遗传学改变,以阐明关键的生物学过程。方法:选择LUAD肿瘤冷冻切片作为研究模型,其中既有侵袭性亚型,也有非侵袭性亚型。这些亚型被鉴定,显微解剖,并单独处理RNA测序(RNA-seq)。随后的分析确定了差异表达基因(DEGs)和显著富集的生物过程。此外,对独立LUAD组织的RNA-seq数据进行分析以筛选关键基因,并进行H3K27ac染色质免疫沉淀测序(ChIP-seq)以探索潜在的调控机制。最后,使用癌症基因组图谱(TCGA)数据库进行生存分析,以验证所鉴定基因的临床相关性。结果:亚型特异性RNA-seq分析显示,细胞外基质(ECM)的改变是LUAD侵袭的一个关键标志,这一点通过独立组织样本的转录组学变化得到了验证。此外,这些ECM基因中的一些与LUAD预后显著相关。基于这些发现,我们假设LUAD进展过程中的表观遗传改变可能驱动这些ECM变化,随后免疫微环境的重塑也可能有助于侵袭过程。结论:我们的综合分析表明,表观遗传和转录组失调诱导的ECM改变是LUAD侵袭的关键。具体来说,关键的ECM基因,包括AGER和CGNL1,被确定为侵袭的中枢调节因子,因此代表了LUAD有希望的治疗靶点。
{"title":"Subtype-specific RNA sequencing using micro-dissection revealed extracellular matrix alterations as key factors in lung adenocarcinoma invasion.","authors":"Chongze Yuan, Xiao Chen, Yizhou Peng, Qiang Zheng, Yunjian Pan, Xuxia Shen, Xiaoting Tao, Xingxin Yao, Hui Hong, Hongbin Ji, Yawei Zhang, Yihua Sun","doi":"10.21037/tlcr-2025-520","DOIUrl":"10.21037/tlcr-2025-520","url":null,"abstract":"<p><strong>Background: </strong>Tumor invasion is a critical step in tumorigenesis and represents an important therapeutic target. However, the molecular mechanisms underlying the invasion process of lung adenocarcinoma (LUAD) remain poorly understood. In this study, we investigated the transcriptomic and epigenetic alterations occurring during LUAD invasion to elucidate the key biological processes.</p><p><strong>Methods: </strong>Frozen section of LUAD tumors, which contained both invasive and non-invasive subtypes, was selected as the study model. These subtypes were identified, micro-dissected, and separately processed for RNA sequencing (RNA-seq). Subsequent analysis identified differentially expressed genes (DEGs) and significantly enriched biological processes. Additionally, RNA-seq data from independent LUAD tissues were analyzed to screen for critical genes, and H3K27ac chromatin immunoprecipitation sequencing (ChIP-seq) was conducted to explore potential regulatory mechanisms. Finally, survival analysis was performed using The Cancer Genome Atlas (TCGA) database to validate the clinical relevance of the identified genes.</p><p><strong>Results: </strong>Subtype-specific RNA-seq analysis revealed that alteration in the extracellular matrix (ECM) was a key hallmark of LUAD invasion, which was validated by transcriptomic changes in independent tissue samples. Furthermore, several of these ECM genes were significantly associated with LUAD prognosis. Based on these findings, we hypothesized that epigenetic alterations during LUAD progression may drive these ECM changes, and that subsequent remodeling of the immune microenvironment may also contribute to the invasive process.</p><p><strong>Conclusions: </strong>Our integrated analysis demonstrated that epigenetic and transcriptomic dysregulation-induced ECM alterations were critical for LUAD invasion. Specifically, key ECM genes, including <i>AGER</i> and <i>CGNL1</i>, were identified as central regulators of invasion and thus represent promising therapeutic targets for LUAD.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"4784-4795"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683414/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715899","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
Stage- and smoking-associated microRNA expression in lung adenocarcinoma. 肺腺癌分期和吸烟相关的microRNA表达。
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-11-30 Epub Date: 2025-11-27 DOI: 10.21037/tlcr-2025-636
Min Huang, Yimin Ge, Huiqin Chen, Caimiao Wei, David Cogdell, Cristina Ivan, Meng Chen, Wei Zhang, George A Calin, Ming Guo

Background: Altered expression of microRNAs (miRNAs) is implicated in lung carcinogenesis, but little research has investigated the association of miRNA alterations with lung cancer stage or smoking status. To identify such alterations in lung adenocarcinoma, we conducted miRNA profiling.

Methods: Lung adenocarcinoma specimens and paired nonneoplastic lung tissues from 58 patients who had received no preoperative therapy and underwent tumor resection from 1991 to 2006 were collected from tumor tissue banks. Thirty (52%) of the tumors were stage I and 28 (48%) stage II or higher. Twenty-five (43%) patients were nonsmokers and 33 (57%) were smokers. To identify miRNAs of which its expression was associated with disease stage and/or smoking status, we performed subgroup analyses based on disease stage and smoking status, alongside overall profiling of all samples. miRNA expression was profiled using a microarray kit in tumors and paired nonneoplastic lung tissues. We assessed the fold changes (FCs) and local false discovery rates (FDRs) for the intercept in addition to P values.

Results: Of the 706 miRNAs examined, 64 had overall altered expression. Of these, 36 (56%) were associated with all stages of adenocarcinoma in both smokers and nonsmokers. In stage-based analysis, 20 altered miRNAs were associated with stage I disease and 38 with stage II or higher disease. Of the 20 miRNAs associated with stage I adenocarcinoma, 4 were miR-200 family members, and miR-200b was the most highly upregulated. Analysis based on smoking status identified 38 altered miRNAs in nonsmokers and 6 altered miRNAs in smokers. Three miR-200 family members were significantly altered in nonsmokers. Among the miRNA alterations associated with stage I adenocarcinoma in nonsmokers, we found alterations in all 5 miR-200 family members (miR-200b, miR-200a, miR-141, miR-429, and miR-200c).

Conclusions: Our identification of miRNA alterations associated with early-stage lung adenocarcinoma in nonsmokers, especially alterations in the miR-200 family, suggests that these miRNAs may play a unique role in the early stages of lung carcinogenesis and progression in nonsmokers and that they may be useful as markers for the early detection of lung cancer.

背景:microrna (miRNAs)表达改变与肺癌发生有关,但很少有研究调查miRNA改变与肺癌分期或吸烟状况的关系。为了确定肺腺癌中的这种改变,我们进行了miRNA谱分析。方法:从1991 ~ 2006年肿瘤组织库中收集58例术前未接受治疗并行肿瘤切除术的患者肺腺癌标本及配对非肿瘤性肺组织。30例(52%)为I期,28例(48%)为II期及以上。25例(43%)患者不吸烟,33例(57%)患者吸烟。为了确定其表达与疾病分期和/或吸烟状况相关的mirna,我们根据疾病分期和吸烟状况进行了亚组分析,并对所有样本进行了总体分析。使用微阵列试剂盒分析miRNA在肿瘤和配对的非肿瘤性肺组织中的表达。除了P值外,我们还评估了截距的折叠变化(FCs)和局部错误发现率(FDRs)。结果:在检测的706个mirna中,64个整体表达改变。其中,36例(56%)与吸烟者和非吸烟者的所有阶段腺癌相关。在基于分期的分析中,20个改变的mirna与I期疾病相关,38个与II期或更高阶段的疾病相关。在与I期腺癌相关的20个mirna中,有4个是miR-200家族成员,miR-200b是上调幅度最大的。基于吸烟状况的分析发现,非吸烟者中有38个改变的mirna,吸烟者中有6个改变的mirna。在不吸烟者中,三个miR-200家族成员显著改变。在与非吸烟者I期腺癌相关的miRNA改变中,我们发现所有5个miR-200家族成员(miR-200b、miR-200a、miR-141、miR-429和miR-200c)都发生了改变。结论:我们鉴定的与非吸烟者早期肺腺癌相关的miRNA改变,特别是miR-200家族的改变,表明这些miRNA可能在非吸烟者早期肺癌发生和进展中发挥独特作用,并且它们可能作为肺癌早期检测的标记物。
{"title":"Stage- and smoking-associated microRNA expression in lung adenocarcinoma.","authors":"Min Huang, Yimin Ge, Huiqin Chen, Caimiao Wei, David Cogdell, Cristina Ivan, Meng Chen, Wei Zhang, George A Calin, Ming Guo","doi":"10.21037/tlcr-2025-636","DOIUrl":"10.21037/tlcr-2025-636","url":null,"abstract":"<p><strong>Background: </strong>Altered expression of microRNAs (miRNAs) is implicated in lung carcinogenesis, but little research has investigated the association of miRNA alterations with lung cancer stage or smoking status. To identify such alterations in lung adenocarcinoma, we conducted miRNA profiling.</p><p><strong>Methods: </strong>Lung adenocarcinoma specimens and paired nonneoplastic lung tissues from 58 patients who had received no preoperative therapy and underwent tumor resection from 1991 to 2006 were collected from tumor tissue banks. Thirty (52%) of the tumors were stage I and 28 (48%) stage II or higher. Twenty-five (43%) patients were nonsmokers and 33 (57%) were smokers. To identify miRNAs of which its expression was associated with disease stage and/or smoking status, we performed subgroup analyses based on disease stage and smoking status, alongside overall profiling of all samples. miRNA expression was profiled using a microarray kit in tumors and paired nonneoplastic lung tissues. We assessed the fold changes (FCs) and local false discovery rates (FDRs) for the intercept in addition to P values.</p><p><strong>Results: </strong>Of the 706 miRNAs examined, 64 had overall altered expression. Of these, 36 (56%) were associated with all stages of adenocarcinoma in both smokers and nonsmokers. In stage-based analysis, 20 altered miRNAs were associated with stage I disease and 38 with stage II or higher disease. Of the 20 miRNAs associated with stage I adenocarcinoma, 4 were miR-200 family members, and miR-200b was the most highly upregulated. Analysis based on smoking status identified 38 altered miRNAs in nonsmokers and 6 altered miRNAs in smokers. Three miR-200 family members were significantly altered in nonsmokers. Among the miRNA alterations associated with stage I adenocarcinoma in nonsmokers, we found alterations in all 5 miR-200 family members (miR-200b, miR-200a, miR-141, miR-429, and miR-200c).</p><p><strong>Conclusions: </strong>Our identification of miRNA alterations associated with early-stage lung adenocarcinoma in nonsmokers, especially alterations in the miR-200 family, suggests that these miRNAs may play a unique role in the early stages of lung carcinogenesis and progression in nonsmokers and that they may be useful as markers for the early detection of lung cancer.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"4942-4961"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683391/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715808","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
Robotic-assisted and electromagnetic navigation bronchoscopy for multi-focal lung cancers: a narrative review. 机器人辅助和电磁导航支气管镜治疗多灶性肺癌的综述。
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-11-30 Epub Date: 2025-11-25 DOI: 10.21037/tlcr-2025-943
Wei Liu, Aliss T C Chang, Joyce W Y Chan, Rainbow W H Lau, Tony S K Mok, Calvin S H Ng

Background and objective: The incidence of multi-focal lung cancers (MFLCs) has increased with the widespread use of low-dose computed tomography, posing significant diagnostic and therapeutic challenges, particularly in differentiating multiple primary lung cancers from intrapulmonary metastasis. Surgical resection remains the standard treatment for early localized disease; however, it is often unsuitable for patients with multi-focal tumors, lesions near vital structures, or impaired lung function. This review aims to summarize current evidence on the use of minimally invasive bronchoscopic platforms, including robotic-assisted bronchoscopy (RAB) and electromagnetic navigation bronchoscopy (ENB), in the diagnosis and treatment of MFLCs.

Methods: A literature search of PubMed, Embase, and Web of Science was performed with a primary search on February 28, 2025 and a final update on September 30, 2025; only English-language publications were included. Keywords included "multi-focal lung cancer", "robotic-assisted bronchoscopy", "electromagnetic navigation bronchoscopy", "bronchoscopic ablation", and related terms. Relevant original studies, clinical trials, case series, reviews, and abstracts were included.

Key content and findings: RAB and ENB enable precise localization and access to peripheral lung lesions, enabling diagnostic biopsy and bronchoscopic ablation while preserving lung tissue. These approaches are particularly advantageous for patients who are poor surgical candidates, allowing the treatment of multiple lesions in a single session with shorter recovery times. Integration with stereotactic body radiation therapy, immunotherapy, and molecular profiling may further enhance local control and personalize therapy.

Conclusions: Bronchoscopic navigation platforms represent a promising addition to multidisciplinary MFLCs management by broadening eligibility for minimally invasive treatment and preserving lung function. With continued refinement and incorporation into multimodal strategies, these technologies may reshape therapeutic algorithms and influence future clinical practice and research direction.

背景和目的:随着低剂量计算机断层扫描的广泛使用,多灶性肺癌(MFLCs)的发病率增加,这给诊断和治疗带来了重大挑战,特别是在区分多发性原发性肺癌和肺内转移方面。手术切除仍然是早期局限性疾病的标准治疗方法;然而,它通常不适用于多灶性肿瘤、重要结构附近病变或肺功能受损的患者。本综述旨在总结微创支气管镜平台在MFLCs诊断和治疗中的应用,包括机器人辅助支气管镜(RAB)和电磁导航支气管镜(ENB)。方法:对PubMed、Embase和Web of Science进行文献检索,首次检索时间为2025年2月28日,最终更新时间为2025年9月30日;只包括英文出版物。关键词包括“多灶性肺癌”、“机器人辅助支气管镜检查”、“电磁导航支气管镜检查”、“支气管镜消融”及相关术语。包括相关的原始研究、临床试验、病例系列、综述和摘要。关键内容和发现:RAB和ENB能够精确定位和进入周围肺病变,在保留肺组织的同时实现诊断活检和支气管镜消融。这些方法对手术条件差的患者特别有利,可以在一次治疗中治疗多个病变,恢复时间更短。结合立体定向放射治疗、免疫治疗和分子谱分析可以进一步加强局部控制和个性化治疗。结论:支气管镜导航平台通过扩大微创治疗的资格和保留肺功能,代表了多学科MFLCs管理的一个有希望的补充。随着多模式策略的不断完善和整合,这些技术可能会重塑治疗算法,并影响未来的临床实践和研究方向。
{"title":"Robotic-assisted and electromagnetic navigation bronchoscopy for multi-focal lung cancers: a narrative review.","authors":"Wei Liu, Aliss T C Chang, Joyce W Y Chan, Rainbow W H Lau, Tony S K Mok, Calvin S H Ng","doi":"10.21037/tlcr-2025-943","DOIUrl":"10.21037/tlcr-2025-943","url":null,"abstract":"<p><strong>Background and objective: </strong>The incidence of multi-focal lung cancers (MFLCs) has increased with the widespread use of low-dose computed tomography, posing significant diagnostic and therapeutic challenges, particularly in differentiating multiple primary lung cancers from intrapulmonary metastasis. Surgical resection remains the standard treatment for early localized disease; however, it is often unsuitable for patients with multi-focal tumors, lesions near vital structures, or impaired lung function. This review aims to summarize current evidence on the use of minimally invasive bronchoscopic platforms, including robotic-assisted bronchoscopy (RAB) and electromagnetic navigation bronchoscopy (ENB), in the diagnosis and treatment of MFLCs.</p><p><strong>Methods: </strong>A literature search of PubMed, Embase, and Web of Science was performed with a primary search on February 28, 2025 and a final update on September 30, 2025; only English-language publications were included. Keywords included \"multi-focal lung cancer\", \"robotic-assisted bronchoscopy\", \"electromagnetic navigation bronchoscopy\", \"bronchoscopic ablation\", and related terms. Relevant original studies, clinical trials, case series, reviews, and abstracts were included.</p><p><strong>Key content and findings: </strong>RAB and ENB enable precise localization and access to peripheral lung lesions, enabling diagnostic biopsy and bronchoscopic ablation while preserving lung tissue. These approaches are particularly advantageous for patients who are poor surgical candidates, allowing the treatment of multiple lesions in a single session with shorter recovery times. Integration with stereotactic body radiation therapy, immunotherapy, and molecular profiling may further enhance local control and personalize therapy.</p><p><strong>Conclusions: </strong>Bronchoscopic navigation platforms represent a promising addition to multidisciplinary MFLCs management by broadening eligibility for minimally invasive treatment and preserving lung function. With continued refinement and incorporation into multimodal strategies, these technologies may reshape therapeutic algorithms and influence future clinical practice and research direction.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"5159-5180"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683444/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715842","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
A public data-based molecular classification of small cell lung cancer by neuroactive signaling networks unveils distinct microenvironment landscapes and immunotherapy-related prognostic biomarkers. 一项基于公共数据的小细胞肺癌分子分类,通过神经活性信号网络揭示了不同的微环境景观和免疫治疗相关的预后生物标志物。
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-11-30 Epub Date: 2025-11-27 DOI: 10.21037/tlcr-2025-620
Wensheng Zhou, Yujie Tang, Jiyuan Zeng, Xiaoyi Zhang, Haotian Meng, Wenhui Guan, Yue Zhu, Huixin Jiang, Yansheng Wang, Xiaohong Xie, Chengzhi Zhou, Ming Liu

Background: Small cell lung cancer (SCLC) represents an aggressive malignancy characterized by marked heterogeneity and neuroendocrine differentiation. Despite its clinical significance, the functional landscape of neuroendocrine function, while neuroactive-signaling-related genes (NRGs) in SCLC pathogenesis remains poorly characterized. Therefore, the aim of this study is to classify SCLC based on neuroactive signaling networks and to analyze the characteristics of these classifications in relation to the immune microenvironment.

Methods: Through integrated analysis of bulk transcriptomic profiling from 79 primary SCLC tumors and single-cell transcriptomic profiling from 11 SCLC tumors, we employed a consensus clustering algorithm to deconvolute transcriptional programs underlying neuroactive signaling networks. Molecular functions and tumor-infiltrated immune cells were estimated from bulk transcriptomes using bioinformatics methods. Single-cell transcriptomic analysis was implemented for cross-validation and cellular characterization.

Results: Bulk-seq analyses reported that the transcriptional variability of three major clusters of tumors were associated with different clinical outcomes and biological pathways. Clinical, genomic, and immunological characteristics were observed among three clusters. Furthermore, the key genes module of cluster with the worst survival were identified as neuroactive-signaling-related signature (NRS) and used to classify tumor samples into two distinct intra-tumoral subtypes (H-NRS and L-NRS) with single-cell transcriptomic data. At single-cell level, malignant cells in H-NRS tumor were in later cell state and had more frequent cellular communication. And NRS subsequently was identified as a biomarker correlated with better prognosis for patients receiving chemoimmunotherapy. It was found that Natriuretic Peptide C (NPPC), as one of the key genes in NRS, was overexpressed in SCLC tumor cells and correlated with poor prognosis. Treatment with C-type natriuretic peptide (CNP) facilitates cellular migration and metastatic potential.

Conclusions: This study proposes a novel molecular taxonomy for SCLC grounded in neuroactive signaling networks, suggests a potential prognostic biomarker to aid in therapeutic stratification, and identifies NPPC as a candidate therapeutic target worthy of further investigation in metastatic SCLC. Our findings may help bridge gaps in understanding between neuroendocrine biology and tumor microenvironment (TME) dynamics during SCLC evolution.

背景:小细胞肺癌(SCLC)是一种侵袭性恶性肿瘤,具有明显的异质性和神经内分泌分化。尽管具有临床意义,但神经内分泌功能的功能景观,以及神经活性信号相关基因(NRGs)在SCLC发病机制中的作用仍不清楚。因此,本研究的目的是基于神经活动信号网络对SCLC进行分类,并分析这些分类的特征与免疫微环境的关系。方法:通过对79例原发性SCLC肿瘤的整体转录组谱和11例SCLC肿瘤的单细胞转录组谱进行综合分析,我们采用共识聚类算法来解卷积神经活性信号网络背后的转录程序。利用生物信息学方法从大量转录组中估计分子功能和肿瘤浸润免疫细胞。单细胞转录组分析用于交叉验证和细胞表征。结果:Bulk-seq分析报告了三种主要肿瘤簇的转录变异性与不同的临床结局和生物学途径相关。临床,基因组学和免疫学特征观察三个集群。此外,生存最差的集群的关键基因模块被确定为神经活性信号相关特征(NRS),并使用单细胞转录组数据将肿瘤样本分为两种不同的肿瘤内亚型(H-NRS和L-NRS)。在单细胞水平上,H-NRS肿瘤中的恶性细胞处于较晚的细胞状态,细胞间通讯更为频繁。NRS随后被确定为与接受化学免疫治疗的患者预后良好相关的生物标志物。研究发现,钠肽C (NPPC)作为NRS的关键基因之一,在SCLC肿瘤细胞中过表达,与不良预后相关。用c型利钠肽(CNP)治疗可促进细胞迁移和转移潜能。结论:本研究提出了一种基于神经活动信号网络的SCLC新的分子分类方法,提出了一种潜在的预后生物标志物来帮助治疗分层,并确定了NPPC作为转移性SCLC的候选治疗靶点,值得进一步研究。我们的研究结果可能有助于弥合SCLC进化过程中神经内分泌生物学和肿瘤微环境(TME)动力学之间的空白。
{"title":"A public data-based molecular classification of small cell lung cancer by neuroactive signaling networks unveils distinct microenvironment landscapes and immunotherapy-related prognostic biomarkers.","authors":"Wensheng Zhou, Yujie Tang, Jiyuan Zeng, Xiaoyi Zhang, Haotian Meng, Wenhui Guan, Yue Zhu, Huixin Jiang, Yansheng Wang, Xiaohong Xie, Chengzhi Zhou, Ming Liu","doi":"10.21037/tlcr-2025-620","DOIUrl":"10.21037/tlcr-2025-620","url":null,"abstract":"<p><strong>Background: </strong>Small cell lung cancer (SCLC) represents an aggressive malignancy characterized by marked heterogeneity and neuroendocrine differentiation. Despite its clinical significance, the functional landscape of neuroendocrine function, while neuroactive-signaling-related genes (NRGs) in SCLC pathogenesis remains poorly characterized. Therefore, the aim of this study is to classify SCLC based on neuroactive signaling networks and to analyze the characteristics of these classifications in relation to the immune microenvironment.</p><p><strong>Methods: </strong>Through integrated analysis of bulk transcriptomic profiling from 79 primary SCLC tumors and single-cell transcriptomic profiling from 11 SCLC tumors, we employed a consensus clustering algorithm to deconvolute transcriptional programs underlying neuroactive signaling networks. Molecular functions and tumor-infiltrated immune cells were estimated from bulk transcriptomes using bioinformatics methods. Single-cell transcriptomic analysis was implemented for cross-validation and cellular characterization.</p><p><strong>Results: </strong>Bulk-seq analyses reported that the transcriptional variability of three major clusters of tumors were associated with different clinical outcomes and biological pathways. Clinical, genomic, and immunological characteristics were observed among three clusters. Furthermore, the key genes module of cluster with the worst survival were identified as neuroactive-signaling-related signature (NRS) and used to classify tumor samples into two distinct intra-tumoral subtypes (H-NRS and L-NRS) with single-cell transcriptomic data. At single-cell level, malignant cells in H-NRS tumor were in later cell state and had more frequent cellular communication. And NRS subsequently was identified as a biomarker correlated with better prognosis for patients receiving chemoimmunotherapy. It was found that Natriuretic Peptide C (NPPC), as one of the key genes in NRS, was overexpressed in SCLC tumor cells and correlated with poor prognosis. Treatment with C-type natriuretic peptide (CNP) facilitates cellular migration and metastatic potential.</p><p><strong>Conclusions: </strong>This study proposes a novel molecular taxonomy for SCLC grounded in neuroactive signaling networks, suggests a potential prognostic biomarker to aid in therapeutic stratification, and identifies NPPC as a candidate therapeutic target worthy of further investigation in metastatic SCLC. Our findings may help bridge gaps in understanding between neuroendocrine biology and tumor microenvironment (TME) dynamics during SCLC evolution.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"4983-4999"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683401/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715869","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
Establishment and characterization of an orthotopic implanted lung cancer model to mimic human tumor structure, microenvironment, and metastatic spread. 模拟人类肿瘤结构、微环境和转移扩散的原位移植肺癌模型的建立和表征。
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-11-30 Epub Date: 2025-11-25 DOI: 10.21037/tlcr-2025-871
Beñat Picabea, Daniel Orive, Covadonga Rodríguez, Miren Mailharin, María Sangüesa, Maeva Houry, Andrea Arricibita, Mirari Echepare, Ane Álava, Cristina Viu-Idocin, Alfonso Calvo, Joaquín Fernández-Irigoyen, Enrique Santamaría, Mikel Ariz, Luis M Montuenga, Karmele Valencia

Background: Subcutaneous (SC) lung tumor models are widely used in preclinical studies due to their technical simplicity but fail to recapitulate the complex microenvironment, immune landscape, and metastatic behavior of human lung cancers. These limitations hinder the translational value of such models, particularly in evaluating immunotherapies and metastasis-related mechanisms. There is a critical need for more physiologically relevant in vivo models that better reflect clinical tumor characteristics and disease progression. To address these limitations, we sought to develop a reproducible orthotopic lung cancer (LuO) model that enables detailed study of tumor progression, immune infiltration, and metastatic dynamics.

Methods: We established and characterized a thoracotomy-based LuO model using a panel of human and murine lung cancer cell lines implanted into the pulmonary parenchyma of immunodeficient and syngeneic mice. Tumor progression was monitored longitudinally using bioluminescence imaging (BLI) and micro-computed tomography (CT). Comparative analyses with SC tumors were performed using immunohistochemistry, multiplexed immunofluorescence, transcriptomic and proteomic analyses. Circulating tumor cells (CTCs) and spontaneous metastases were isolated and functionally characterized.

Results: The orthotopic model reliably generated solitary intrapulmonary tumors that closely mimic human lung cancer in growth pattern, vascularization, and progression. Compared to SC tumors, orthotopic tumors exhibited significantly enhanced vascular density, reduced hypoxia and DNA damage, and increased proliferation. Immune profiling revealed enriched and spatially organized infiltrates of CD4+, CD8+ T cells, dendritic cells (DCs), and myeloid populations in orthotopic tumors, forming structures analogous to those found in patient tumors. Moreover, orthotopic tumors released CTCs capable of forming spontaneous and site-specific metastases to clinically relevant organs. Transcriptomic and proteomic profiling of metastasis-derived cell lines uncovered conserved pro-metastatic signatures and niche-specific adaptations.

Conclusions: This LuO model offers a reproducible, clinically relevant platform that captures important aspects of human lung cancer biology, including immune landscape, tumor microenvironment (TME), and metastatic progression. Its superior anatomical and immunological fidelity makes it a valuable preclinical tool for evaluating therapeutic strategies and dissecting molecular mechanisms of metastasis.

背景:皮下(SC)肺肿瘤模型由于其技术简单而广泛用于临床前研究,但未能概括人类肺癌复杂的微环境,免疫景观和转移行为。这些限制阻碍了这些模型的转化价值,特别是在评估免疫疗法和转移相关机制方面。迫切需要更多与生理相关的体内模型,以更好地反映临床肿瘤特征和疾病进展。为了解决这些局限性,我们试图建立一个可重复的原位肺癌(LuO)模型,该模型可以详细研究肿瘤进展、免疫浸润和转移动力学。方法:将人、鼠肺癌细胞系移植到免疫缺陷小鼠和同基因小鼠肺实质中,建立一种基于开胸的LuO模型,并对其进行表征。采用生物发光成像(BLI)和微型计算机断层扫描(CT)纵向监测肿瘤进展。采用免疫组织化学、多重免疫荧光、转录组学和蛋白质组学分析与SC肿瘤进行比较分析。循环肿瘤细胞(ctc)和自发转移瘤被分离出来并进行功能表征。结果:原位模型可靠地生成孤立的肺内肿瘤,其生长模式、血管形成和进展与人类肺癌非常相似。与SC肿瘤相比,原位肿瘤血管密度明显增强,缺氧和DNA损伤减少,增殖增加。免疫图谱显示,在原位肿瘤中,CD4+、CD8+ T细胞、树突状细胞(dc)和髓系细胞群丰富且有空间组织浸润,形成与患者肿瘤相似的结构。此外,原位肿瘤释放的ctc能够形成自发和部位特异性转移到临床相关器官。转移来源细胞系的转录组学和蛋白质组学分析揭示了保守的前转移特征和小生境特异性适应。结论:该LuO模型提供了一个可重复的、临床相关的平台,可以捕获人类肺癌生物学的重要方面,包括免疫景观、肿瘤微环境(TME)和转移进展。其优越的解剖学和免疫学保真度使其成为评估治疗策略和解剖转移分子机制的有价值的临床前工具。
{"title":"Establishment and characterization of an orthotopic implanted lung cancer model to mimic human tumor structure, microenvironment, and metastatic spread.","authors":"Beñat Picabea, Daniel Orive, Covadonga Rodríguez, Miren Mailharin, María Sangüesa, Maeva Houry, Andrea Arricibita, Mirari Echepare, Ane Álava, Cristina Viu-Idocin, Alfonso Calvo, Joaquín Fernández-Irigoyen, Enrique Santamaría, Mikel Ariz, Luis M Montuenga, Karmele Valencia","doi":"10.21037/tlcr-2025-871","DOIUrl":"10.21037/tlcr-2025-871","url":null,"abstract":"<p><strong>Background: </strong>Subcutaneous (SC) lung tumor models are widely used in preclinical studies due to their technical simplicity but fail to recapitulate the complex microenvironment, immune landscape, and metastatic behavior of human lung cancers. These limitations hinder the translational value of such models, particularly in evaluating immunotherapies and metastasis-related mechanisms. There is a critical need for more physiologically relevant <i>in vivo</i> models that better reflect clinical tumor characteristics and disease progression. To address these limitations, we sought to develop a reproducible orthotopic lung cancer (LuO) model that enables detailed study of tumor progression, immune infiltration, and metastatic dynamics.</p><p><strong>Methods: </strong>We established and characterized a thoracotomy-based LuO model using a panel of human and murine lung cancer cell lines implanted into the pulmonary parenchyma of immunodeficient and syngeneic mice. Tumor progression was monitored longitudinally using bioluminescence imaging (BLI) and micro-computed tomography (CT). Comparative analyses with SC tumors were performed using immunohistochemistry, multiplexed immunofluorescence, transcriptomic and proteomic analyses. Circulating tumor cells (CTCs) and spontaneous metastases were isolated and functionally characterized.</p><p><strong>Results: </strong>The orthotopic model reliably generated solitary intrapulmonary tumors that closely mimic human lung cancer in growth pattern, vascularization, and progression. Compared to SC tumors, orthotopic tumors exhibited significantly enhanced vascular density, reduced hypoxia and DNA damage, and increased proliferation. Immune profiling revealed enriched and spatially organized infiltrates of CD4<sup>+</sup>, CD8<sup>+</sup> T cells, dendritic cells (DCs), and myeloid populations in orthotopic tumors, forming structures analogous to those found in patient tumors. Moreover, orthotopic tumors released CTCs capable of forming spontaneous and site-specific metastases to clinically relevant organs. Transcriptomic and proteomic profiling of metastasis-derived cell lines uncovered conserved pro-metastatic signatures and niche-specific adaptations.</p><p><strong>Conclusions: </strong>This LuO model offers a reproducible, clinically relevant platform that captures important aspects of human lung cancer biology, including immune landscape, tumor microenvironment (TME), and metastatic progression. Its superior anatomical and immunological fidelity makes it a valuable preclinical tool for evaluating therapeutic strategies and dissecting molecular mechanisms of metastasis.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"4868-4895"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683413/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715845","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
Real-world first-line outcomes of alectinib and brigatinib in anaplastic lymphoma kinase-positive non-small cell lung cancer: a nationwide South Korean cohort study using the health insurance review and assessment data. 阿勒替尼和布加替尼治疗间变性淋巴瘤激酶阳性非小细胞肺癌的实际一线疗效:一项使用健康保险审查和评估数据的韩国全国性队列研究
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-11-30 Epub Date: 2025-11-27 DOI: 10.21037/tlcr-2025-983
Hee Jun Kim, Eunyoung Angela Lee, Jin-Hee Park, Hyun Woo Lee

Background: Lung cancer remains the leading cause of cancer-related mortality worldwide. Anaplastic lymphoma kinase (ALK)-positive non-small cell lung cancer (NSCLC) presents unique clinical challenges, including frequent central nervous system (CNS) metastases. At present, comparative real-world data on ALK inhibitors remain limited. This study aimed to compare real-world progression-free survival (PFS) and overall survival (OS) among patients with ALK-positive NSCLC treated with first-line alectinib or brigatinib using nationwide South Korean data.

Methods: This retrospective cohort study analyzed anonymized data from South Korea's Health Insurance Review and Assessment Service, covering January 2007 to December 2023. Patients diagnosed with ALK-positive NSCLC (ICD-10: C34x) and treated with either alectinib or brigatinib as first-line monotherapy were included. Patients with prior lung surgery or other malignancies were excluded. Baseline demographics and comorbidities were collected. The primary outcomes were PFS and OS, as measured from ALK inhibitor initiation.

Results: The final cohort included 1,009 patients with ALK-positive NSCLC. The mean age was 61.6 years, and 49.5% were male. Alectinib was associated with a significantly longer PFS. Brigatinib showed a higher OS in the unadjusted analysis; however, this difference was not statistically significant after multivariable adjustment. Transition to lorlatinib was associated with extended survival in both groups, reflecting its use as a later-line therapy following resistance.

Conclusions: In this real-world cohort of ALK-positive NSCLC patients, both alectinib and brigatinib were associated with extended survival, with alectinib showing longer PFS. Findings should be interpreted descriptively. Alectinib demonstrated superior disease control in terms of PFS. Further research is warranted to optimize treatment sequence strategies for ALK inhibitors.

背景:肺癌仍然是世界范围内癌症相关死亡的主要原因。间变性淋巴瘤激酶(ALK)阳性的非小细胞肺癌(NSCLC)具有独特的临床挑战,包括频繁的中枢神经系统(CNS)转移。目前,关于ALK抑制剂的比较真实数据仍然有限。该研究旨在比较使用韩国全国数据的一线alk阳性NSCLC患者的无进展生存期(PFS)和总生存期(OS)。方法:本回顾性队列研究分析了2007年1月至2023年12月韩国健康保险审查和评估服务的匿名数据。诊断为alk阳性NSCLC (ICD-10: C34x)的患者接受阿勒替尼或布加替尼作为一线单药治疗。既往有肺部手术或其他恶性肿瘤的患者被排除在外。收集基线人口统计数据和合并症。主要结局是PFS和OS,从ALK抑制剂开始测量。结果:最终队列包括1009例alk阳性NSCLC患者。平均年龄61.6岁,男性49.5%。Alectinib与更长的PFS相关。布加替尼在未调整分析中显示更高的OS;但经多变量调整后,差异无统计学意义。过渡到氯拉替尼与两组的延长生存期相关,反映了其作为耐药后的后期治疗的使用。结论:在这个真实世界的alk阳性NSCLC患者队列中,阿勒替尼和布加替尼都与延长生存期相关,其中阿勒替尼显示出更长的PFS。应描述性地解释调查结果。在PFS方面,Alectinib显示出优越的疾病控制。需要进一步的研究来优化ALK抑制剂的治疗顺序策略。
{"title":"Real-world first-line outcomes of alectinib and brigatinib in anaplastic lymphoma kinase-positive non-small cell lung cancer: a nationwide South Korean cohort study using the health insurance review and assessment data.","authors":"Hee Jun Kim, Eunyoung Angela Lee, Jin-Hee Park, Hyun Woo Lee","doi":"10.21037/tlcr-2025-983","DOIUrl":"10.21037/tlcr-2025-983","url":null,"abstract":"<p><strong>Background: </strong>Lung cancer remains the leading cause of cancer-related mortality worldwide. Anaplastic lymphoma kinase (ALK)-positive non-small cell lung cancer (NSCLC) presents unique clinical challenges, including frequent central nervous system (CNS) metastases. At present, comparative real-world data on ALK inhibitors remain limited. This study aimed to compare real-world progression-free survival (PFS) and overall survival (OS) among patients with ALK-positive NSCLC treated with first-line alectinib or brigatinib using nationwide South Korean data.</p><p><strong>Methods: </strong>This retrospective cohort study analyzed anonymized data from South Korea's Health Insurance Review and Assessment Service, covering January 2007 to December 2023. Patients diagnosed with ALK-positive NSCLC (ICD-10: C34x) and treated with either alectinib or brigatinib as first-line monotherapy were included. Patients with prior lung surgery or other malignancies were excluded. Baseline demographics and comorbidities were collected. The primary outcomes were PFS and OS, as measured from ALK inhibitor initiation.</p><p><strong>Results: </strong>The final cohort included 1,009 patients with ALK-positive NSCLC. The mean age was 61.6 years, and 49.5% were male. Alectinib was associated with a significantly longer PFS. Brigatinib showed a higher OS in the unadjusted analysis; however, this difference was not statistically significant after multivariable adjustment. Transition to lorlatinib was associated with extended survival in both groups, reflecting its use as a later-line therapy following resistance.</p><p><strong>Conclusions: </strong>In this real-world cohort of ALK-positive NSCLC patients, both alectinib and brigatinib were associated with extended survival, with alectinib showing longer PFS. Findings should be interpreted descriptively. Alectinib demonstrated superior disease control in terms of PFS. Further research is warranted to optimize treatment sequence strategies for ALK inhibitors.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"4811-4823"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683494/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715803","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 structural lipidomic profiling reveals C=C positional isomers as potential biomarkers in lung adenocarcinoma tissue. 深层结构脂质组学分析显示C=C位置异构体是肺腺癌组织中潜在的生物标志物。
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-11-30 Epub Date: 2025-11-27 DOI: 10.21037/tlcr-2025-717
Yang Gu, Ming-Ming Shao, Song-Ping Cui, Bin Hu, Xin Li

Background: Recent evidence highlights the importance of lipid metabolic reprogramming in lung adenocarcinoma (LUAD) progression. Due to the limitations of conventional techniques, the fine structure of lipids cannot be identified. The metabolic changes of lipid structural features-particularly carbon-carbon double bond (C=C) positional isomers-remain underexplored. This study aims to characterize the structural alterations of lipids, especially C=C positional isomers, in LUAD tissues to elucidate their potential roles in tumor progression.

Methods: We performed deep structural lipidomic profiling on paired normal lung (N) and LUAD (T) tissue samples using a combination of photochemical reaction-based structural analysis (Ω Analyzer) and liquid chromatography-mass spectrometry (LC-MS). Lipid species were characterized at three structural levels: lipid class, molecular species, and C=C positional isomer. Relative quantitative analyses were conducted to identify differences in total composition, unsaturation levels, and the distribution of C=C isomers between N and T groups.

Results: A total of 794 phospholipid species were identified at the C=C isomer level, with the T group exhibiting a slightly higher overall number of identified lipids compared to the N group. Polyunsaturated lipids displayed notable upregulation in the T group and facilitated robust clustering between normal and cancer tissues. Furthermore, analyzing C=C positional isomers revealed significant differences in their relative abundances between the two groups: lipids enriched in C18:1(Δ9) were predominantly upregulated in T group samples, whereas those carrying C18:1(Δ8) were generally downregulated.

Conclusions: Our findings demonstrate that deep structural lipidomic analysis yields crucial insights into the lipid reprogramming of LUAD. In particular, the relative abundances of C=C positional isomers hold promise as novel diagnostic markers and therapeutic targets for LUAD.

背景:最近的证据强调了脂质代谢重编程在肺腺癌(LUAD)进展中的重要性。由于传统技术的限制,脂质的精细结构无法被识别。脂质结构特征的代谢变化-特别是碳-碳双键(C=C)位置异构体-仍未得到充分研究。本研究旨在表征LUAD组织中脂质的结构改变,特别是C=C位置异构体,以阐明其在肿瘤进展中的潜在作用。方法:采用基于光化学反应的结构分析(Ω Analyzer)和液相色谱-质谱(LC-MS)相结合的方法对配对的正常肺(N)和LUAD (T)组织样本进行深层结构脂质组学分析。脂类从脂类、分子种类和C=C位置异构体三个结构层次进行表征。通过相关的定量分析来确定N组和T组之间C=C异构体的总组成、不饱和水平和分布的差异。结果:在C=C异构体水平上共鉴定出794种磷脂,其中T组鉴定出的脂质总数略高于N组。多不饱和脂在T组中表现出显著的上调,并促进了正常组织和癌组织之间的强大聚类。此外,分析C=C位置异构体揭示了两组之间相对丰度的显著差异:在T组样品中,富含C18:1(Δ9)的脂质主要上调,而携带C18:1(Δ8)的脂质则普遍下调。结论:我们的研究结果表明,深层结构脂质组学分析对LUAD的脂质重编程产生了至关重要的见解。特别是,C=C位置异构体的相对丰度有望成为LUAD的新诊断标记和治疗靶点。
{"title":"Deep structural lipidomic profiling reveals C=C positional isomers as potential biomarkers in lung adenocarcinoma tissue.","authors":"Yang Gu, Ming-Ming Shao, Song-Ping Cui, Bin Hu, Xin Li","doi":"10.21037/tlcr-2025-717","DOIUrl":"10.21037/tlcr-2025-717","url":null,"abstract":"<p><strong>Background: </strong>Recent evidence highlights the importance of lipid metabolic reprogramming in lung adenocarcinoma (LUAD) progression. Due to the limitations of conventional techniques, the fine structure of lipids cannot be identified. The metabolic changes of lipid structural features-particularly carbon-carbon double bond (C=C) positional isomers-remain underexplored. This study aims to characterize the structural alterations of lipids, especially C=C positional isomers, in LUAD tissues to elucidate their potential roles in tumor progression.</p><p><strong>Methods: </strong>We performed deep structural lipidomic profiling on paired normal lung (N) and LUAD (T) tissue samples using a combination of photochemical reaction-based structural analysis (Ω Analyzer) and liquid chromatography-mass spectrometry (LC-MS). Lipid species were characterized at three structural levels: lipid class, molecular species, and C=C positional isomer. Relative quantitative analyses were conducted to identify differences in total composition, unsaturation levels, and the distribution of C=C isomers between N and T groups.</p><p><strong>Results: </strong>A total of 794 phospholipid species were identified at the C=C isomer level, with the T group exhibiting a slightly higher overall number of identified lipids compared to the N group. Polyunsaturated lipids displayed notable upregulation in the T group and facilitated robust clustering between normal and cancer tissues. Furthermore, analyzing C=C positional isomers revealed significant differences in their relative abundances between the two groups: lipids enriched in C18:1(Δ9) were predominantly upregulated in T group samples, whereas those carrying C18:1(Δ8) were generally downregulated.</p><p><strong>Conclusions: </strong>Our findings demonstrate that deep structural lipidomic analysis yields crucial insights into the lipid reprogramming of LUAD. In particular, the relative abundances of C=C positional isomers hold promise as novel diagnostic markers and therapeutic targets for LUAD.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"4768-4783"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683375/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715511","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
Redefining treatment interval in lung cancer surgery in the era of prehabilitation: a systematic review. 在康复时代重新定义肺癌手术治疗间隔:一项系统综述。
IF 3.5 2区 医学 Q2 ONCOLOGY Pub Date : 2025-11-30 Epub Date: 2025-11-21 DOI: 10.21037/tlcr-2025-688
Lisa D Geomini, Quirine C A van Steenwijk, Shiromani Janki, Iris Hoogendoorn, Gerrit D Slooter, Frank J C van den Broek, Geertruid M H Marres

Background: Time-to-surgery is used as a surrogate quality indicator within lung cancer care. However, its definition is not clearly defined in the literature. This study aimed to explore the evidence on optimal time-to-surgery interval with no negative impact on disease progression or oncological outcomes.

Methods: A systematic search was performed in January 2025 through MEDLINE, EMBASE, and Cochrane databases. Studies about lung cancer patients with treatment interval description until surgery were included.

Results: Eighty-six studies were included with a clear definition of the treatment interval until surgery. Starting points of the interval varied widely, of which pathological diagnosis was reported most often. Thirty-six studies also included associations of time-to-surgery with oncological outcomes. Overall, 47% of the included studies associated a delay in treatment interval with worse outcomes, 33% found no association, and in 19% the association differed per lung cancer stage or type of outcome. Inconsistency was seen in the definition of timely surgery, ranging from 21 to 90 days, influencing the reported outcome measures and comparability. Most studies chose the tipping point for delayed surgery at or beyond 6 weeks, and the most reported outcome was overall survival (OS). For OS, two thirds of the included study groups associated a delay in time-to-surgery with worse survival, with a very heterogeneous cut-off point between timely and delayed surgery.

Conclusions: Within lung cancer care, there is no clear definition of treatment interval until surgery, nor consensus on the definition of timely surgery. The relation between treatment interval and outcomes is inconsistent and requires more structured evidence. We suggest using pathological diagnosis as a starting point, and a 6-week timeframe as a basis to define timely surgery instead of "as soon as possible", without significantly compromising oncological safety. Defining an interval can reframe waiting time into structural preoperative preparation time, unlocking the opportunity to implement prehabilitation and optimize patient outcomes.

背景:手术时间被用作肺癌治疗的替代质量指标。然而,其定义在文献中并没有明确定义。本研究旨在探讨对疾病进展或肿瘤预后无负面影响的最佳手术间隔时间的证据。方法:于2025年1月通过MEDLINE、EMBASE和Cochrane数据库进行系统检索。纳入了有治疗间隔描述的肺癌患者直至手术的研究。结果:86项研究纳入了明确定义的治疗间隔至手术。间隔起始点差异很大,病理诊断报道最多。36项研究还包括手术时间与肿瘤预后的关系。总体而言,47%的纳入研究将治疗间隔的延迟与较差的结果联系起来,33%的研究发现没有关联,19%的研究因肺癌分期或结果类型而异。及时手术的定义不一致,从21天到90天不等,影响了报告的结果测量和可比性。大多数研究选择延迟手术的临界点为6周或6周以上,报道最多的结果是总生存期(OS)。对于OS,三分之二的纳入研究组将延迟手术时间与更差的生存率联系起来,及时手术和延迟手术之间的分界点非常不一致。结论:在肺癌的护理中,对手术前的治疗间隔没有明确的定义,对及时手术的定义也没有共识。治疗间隔和结果之间的关系是不一致的,需要更多的结构化证据。我们建议以病理诊断为出发点,以6周的时间框架为依据,在不明显影响肿瘤安全性的情况下,及时手术而不是“尽快”。定义一个间隔可以将等待时间重新定义为结构化的术前准备时间,为实施康复治疗和优化患者预后创造机会。
{"title":"Redefining treatment interval in lung cancer surgery in the era of prehabilitation: a systematic review.","authors":"Lisa D Geomini, Quirine C A van Steenwijk, Shiromani Janki, Iris Hoogendoorn, Gerrit D Slooter, Frank J C van den Broek, Geertruid M H Marres","doi":"10.21037/tlcr-2025-688","DOIUrl":"10.21037/tlcr-2025-688","url":null,"abstract":"<p><strong>Background: </strong>Time-to-surgery is used as a surrogate quality indicator within lung cancer care. However, its definition is not clearly defined in the literature. This study aimed to explore the evidence on optimal time-to-surgery interval with no negative impact on disease progression or oncological outcomes.</p><p><strong>Methods: </strong>A systematic search was performed in January 2025 through MEDLINE, EMBASE, and Cochrane databases. Studies about lung cancer patients with treatment interval description until surgery were included.</p><p><strong>Results: </strong>Eighty-six studies were included with a clear definition of the treatment interval until surgery. Starting points of the interval varied widely, of which pathological diagnosis was reported most often. Thirty-six studies also included associations of time-to-surgery with oncological outcomes. Overall, 47% of the included studies associated a delay in treatment interval with worse outcomes, 33% found no association, and in 19% the association differed per lung cancer stage or type of outcome. Inconsistency was seen in the definition of timely surgery, ranging from 21 to 90 days, influencing the reported outcome measures and comparability. Most studies chose the tipping point for delayed surgery at or beyond 6 weeks, and the most reported outcome was overall survival (OS). For OS, two thirds of the included study groups associated a delay in time-to-surgery with worse survival, with a very heterogeneous cut-off point between timely and delayed surgery.</p><p><strong>Conclusions: </strong>Within lung cancer care, there is no clear definition of treatment interval until surgery, nor consensus on the definition of timely surgery. The relation between treatment interval and outcomes is inconsistent and requires more structured evidence. We suggest using pathological diagnosis as a starting point, and a 6-week timeframe as a basis to define timely surgery instead of \"as soon as possible\", without significantly compromising oncological safety. Defining an interval can reframe waiting time into structural preoperative preparation time, unlocking the opportunity to implement prehabilitation and optimize patient outcomes.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"5082-5098"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683378/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715811","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