首页 > 最新文献

Translational lung cancer research最新文献

英文 中文
Enhanced tumor targeting and penetration of fluorophores via iRGD peptide conjugation: a strategy for the precision targeting of lung cancer. 通过 iRGD 肽连接增强荧光团的肿瘤靶向性和穿透性:一种肺癌精准靶向治疗策略。
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-08-31 Epub Date: 2024-08-28 DOI: 10.21037/tlcr-24-589
Yunlong Li, Chenmei Li, Jiamin Li, Dong Han, Gang Xu, Daolong Zhu, Huiming Cai, Yiqing Wang, Dong Wang
<p><strong>Background: </strong>Accurate real-time tumor delineation is essential for achieving curative resection (R0 resection) during non-small cell lung cancer (NSCLC) surgery. The unique characteristics of lung tissue structure significantly challenge the use of video-assisted thoracoscopic surgery in the identification of lung nodules. This difficulty often results in an inability to discern the margins of lung nodules, necessitating either an expansion of the resection scope, or a transition to open surgery. Due to its high spatial resolution, ease of operation, and capacity for real-time observation, near-infrared fluorescence (NIRF) navigation in oncological surgery has emerged as a focal point of clinical research. Targeted NIRF probes, which accumulate preferentially in tumor tissues and are rapidly cleared from normal tissues, enhance diagnostic sensitivity and surgical outcomes. The imaging effect of the clinically approved NIRF probe indocyanine green (ICG) varies significantly from person to person. Therefore, we hope to develop a new generation of targeted NIRF probes targeting lung tumor-specific targets.</p><p><strong>Methods: </strong>First, the peptide iRGD (sequence: CRGDKGPDC) fluorescent tracer was synthesized, and characterized through mass spectrometry (MS), proton nuclear magnetic resonance (<sup>1</sup>H NMR), and high-performance liquid chromatography (HPLC). Fluorescence properties were tested subsequently. Safety was performed <i>in vitro</i> using both human normal liver cells and human normal breast cells. Second, Metabolism and optimal imaging time were determined by tail vein injection of iRGD fluorescent tracer. Finally, Orthotopic and metastatic lung tumor models were used to evaluate the targeting properties of the iRGD fluorescent tracer.</p><p><strong>Results: </strong>We successfully synthesized an iRGD fluorescent tracer specifically designed to target NSCLC. The molecular docking analyses indicated that this tracer has receptor affinity comparable to that of iRGD for αvβ3 integrin, with a purity ≥98%. Additionally, the tracer is highly soluble in water, and its excitation and emission wavelengths are 767 and 799 nm, respectively, positioning it within the near-infrared spectrum. The cellular assays confirmed the tracer's minimal cytotoxicity, underscoring its excellent biosafety profile. <i>In vivo</i> studies further validated the tracer's capacity for specific NSCLC detection at the cellular level, alongside a prolonged imaging window of 6 days or more. Notably, the tracer demonstrated superior specificity in localizing very small lung nodules, which are otherwise clinically indiscernible, outperforming non-targeted ICG. Fluorescence intensity analyses across various organs revealed that the tracer is predominantly metabolized by the liver and kidneys, with excretion via bile and urine, and exhibits minimal toxicity to these organs as well as the lungs.</p><p><strong>Conclusions: </strong>The iRGD fluor
背景:在非小细胞肺癌(NSCLC)手术中,准确的实时肿瘤分界对于实现治愈性切除(R0切除)至关重要。肺组织结构的独特性给使用视频辅助胸腔镜手术识别肺结节带来了巨大挑战。这种困难往往导致无法辨别肺结节的边缘,从而不得不扩大切除范围或转为开放手术。由于近红外荧光(NIRF)导航具有空间分辨率高、操作简便、可实时观察等优点,在肿瘤手术中的应用已成为临床研究的重点。有针对性的近红外荧光探针会优先在肿瘤组织中聚集,并迅速从正常组织中清除,从而提高诊断灵敏度和手术效果。临床认可的近红外荧光探针吲哚菁绿(ICG)的成像效果因人而异。因此,我们希望开发出针对肺部肿瘤特异性靶点的新一代靶向近红外荧光探针:首先,合成了多肽 iRGD(序列:CRGDKGPDC)荧光示踪剂,并通过质谱(MS)、质子核磁共振(1H NMR)和高效液相色谱(HPLC)对其进行了表征。随后还测试了荧光特性。在体外使用人类正常肝细胞和人类正常乳腺细胞进行了安全性测试。其次,通过尾静脉注射 iRGD 荧光示踪剂确定了代谢和最佳成像时间。最后,我们利用原位和转移性肺肿瘤模型评估了 iRGD 荧光示踪剂的靶向特性:我们成功合成了一种专为靶向 NSCLC 而设计的 iRGD 荧光示踪剂。分子对接分析表明,该示踪剂对αvβ3整合素的受体亲和力与iRGD相当,纯度≥98%。此外,该示踪剂极易溶于水,其激发和发射波长分别为 767 纳米和 799 纳米,属于近红外光谱。细胞检测证实示踪剂的细胞毒性极低,突出了其出色的生物安全性。体内研究进一步验证了该示踪剂在细胞水平特异性检测 NSCLC 的能力,以及 6 天或更长时间的成像窗口。值得注意的是,该示踪剂在定位非常小的肺部结节(否则临床上无法辨别)方面表现出卓越的特异性,优于非靶向 ICG。各器官的荧光强度分析表明,示踪剂主要由肝脏和肾脏代谢,通过胆汁和尿液排出体外,对这些器官和肺部的毒性极小:iRGD荧光示踪剂通过特异性靶向肿瘤细胞表面过度表达的αvβ3受体,选择性地在NSCLC组织中蓄积。这种靶向方法有助于术中对 NSCLC 进行实时定位,是一种改进的术中肿瘤识别策略,具有巨大的临床应用潜力。
{"title":"Enhanced tumor targeting and penetration of fluorophores via iRGD peptide conjugation: a strategy for the precision targeting of lung cancer.","authors":"Yunlong Li, Chenmei Li, Jiamin Li, Dong Han, Gang Xu, Daolong Zhu, Huiming Cai, Yiqing Wang, Dong Wang","doi":"10.21037/tlcr-24-589","DOIUrl":"https://doi.org/10.21037/tlcr-24-589","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;Accurate real-time tumor delineation is essential for achieving curative resection (R0 resection) during non-small cell lung cancer (NSCLC) surgery. The unique characteristics of lung tissue structure significantly challenge the use of video-assisted thoracoscopic surgery in the identification of lung nodules. This difficulty often results in an inability to discern the margins of lung nodules, necessitating either an expansion of the resection scope, or a transition to open surgery. Due to its high spatial resolution, ease of operation, and capacity for real-time observation, near-infrared fluorescence (NIRF) navigation in oncological surgery has emerged as a focal point of clinical research. Targeted NIRF probes, which accumulate preferentially in tumor tissues and are rapidly cleared from normal tissues, enhance diagnostic sensitivity and surgical outcomes. The imaging effect of the clinically approved NIRF probe indocyanine green (ICG) varies significantly from person to person. Therefore, we hope to develop a new generation of targeted NIRF probes targeting lung tumor-specific targets.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;First, the peptide iRGD (sequence: CRGDKGPDC) fluorescent tracer was synthesized, and characterized through mass spectrometry (MS), proton nuclear magnetic resonance (&lt;sup&gt;1&lt;/sup&gt;H NMR), and high-performance liquid chromatography (HPLC). Fluorescence properties were tested subsequently. Safety was performed &lt;i&gt;in vitro&lt;/i&gt; using both human normal liver cells and human normal breast cells. Second, Metabolism and optimal imaging time were determined by tail vein injection of iRGD fluorescent tracer. Finally, Orthotopic and metastatic lung tumor models were used to evaluate the targeting properties of the iRGD fluorescent tracer.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;We successfully synthesized an iRGD fluorescent tracer specifically designed to target NSCLC. The molecular docking analyses indicated that this tracer has receptor affinity comparable to that of iRGD for αvβ3 integrin, with a purity ≥98%. Additionally, the tracer is highly soluble in water, and its excitation and emission wavelengths are 767 and 799 nm, respectively, positioning it within the near-infrared spectrum. The cellular assays confirmed the tracer's minimal cytotoxicity, underscoring its excellent biosafety profile. &lt;i&gt;In vivo&lt;/i&gt; studies further validated the tracer's capacity for specific NSCLC detection at the cellular level, alongside a prolonged imaging window of 6 days or more. Notably, the tracer demonstrated superior specificity in localizing very small lung nodules, which are otherwise clinically indiscernible, outperforming non-targeted ICG. Fluorescence intensity analyses across various organs revealed that the tracer is predominantly metabolized by the liver and kidneys, with excretion via bile and urine, and exhibits minimal toxicity to these organs as well as the lungs.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Conclusions: &lt;/strong&gt;The iRGD fluor","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11384490/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142296332","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
Research into overcoming drug resistance in lung cancer treatment using CRISPR-Cas9 technology: a narrative review. 利用 CRISPR-Cas9 技术克服肺癌治疗耐药性的研究:综述。
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-08-31 Epub Date: 2024-08-28 DOI: 10.21037/tlcr-24-592
Bin Liu, Ziyu Wang, Meng Gu, Jinghui Wang, Jinjing Tan

Background and objective: Lung cancer remains a leading cause of cancer-related mortality globally, with drug resistance posing a significant challenge to effective treatment. The advent of clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (CRISPR-Cas9) technology offers a novel and precise gene-editing technology for targeting and negating drug resistance mechanisms in lung cancer. This review summarizes the research progress in the use of CRISPR-Cas9 technology for investigating and managing drug resistance in lung cancer treatment.

Methods: A literature search was conducted using the Web of Science and PubMed databases, with the following keywords: [CRISPR-Cas9], [lung cancer], [drug resistance], [gene editing], and [gene therapy]. The search was limited to articles published in English from 2002 to September 2023. From the search results, studies that utilized CRISPR-Cas9 technology in the context of lung cancer drug resistance were selected for further analysis and summarize.

Key content and findings: CRISPR-Cas9 technology enables precise DNA-sequence editing, allowing for the targeted addition, deletion, or modification of genes. It has been applied to investigate drug resistance in lung cancer by focusing on key genes such as epidermal growth factor receptor (EGFR), Kirsten rat sarcoma viral oncogene homolog (KRAS), tumor protein 53 (TP53), and B-cell lymphoma/leukemia-2 (BCL2), among others. The technology has shown potential in inhibiting tumor growth, repairing mutations, and enhancing the sensitivity of cancer cells to chemotherapy. Additionally, CRISPR-Cas9 has been used to identify novel key genes and molecular mechanisms contributing to drug resistance, offering new avenues for therapeutic intervention. The review also highlights the use of CRISPR-Cas9 in targeting immune escape mechanisms and the development of strategies to improve drug sensitivity.

Conclusions: The CRISPR-Cas9 technology holds great promise for advancing lung cancer treatment, particularly in addressing drug resistance. The ability to precisely target and edit genes involved in resistance pathways offers a powerful tool for developing more effective and personalized therapies. While challenges remain in terms of delivery, safety, and ethical considerations, ongoing research and technological refinements are expected to further enhance the role of CRISPR-Cas9 in improving patient outcomes in lung cancer treatment.

背景和目的:肺癌仍然是全球癌症相关死亡的主要原因,其耐药性对有效治疗构成了巨大挑战。聚类规则间隔短回文重复序列(CRISPR)和CRISPR相关蛋白9(CRISPR-Cas9)技术的出现为靶向和消除肺癌耐药机制提供了一种新颖而精确的基因编辑技术。本综述总结了利用CRISPR-Cas9技术研究和管理肺癌治疗耐药性的研究进展:方法:使用 Web of Science 和 PubMed 数据库进行文献检索,关键词如下:[CRISPR-Cas9]、[肺癌]、[耐药性]、[基因编辑]和[基因治疗]。搜索仅限于 2002 年至 2023 年 9 月期间发表的英文文章。从检索结果中筛选出利用 CRISPR-Cas9 技术研究肺癌耐药性的研究进行进一步分析和总结:CRISPR-Cas9技术可实现精确的DNA序列编辑,从而有针对性地添加、删除或修改基因。它已被应用于研究肺癌的耐药性,重点研究表皮生长因子受体(EGFR)、Kirsten大鼠肉瘤病毒癌基因同源物(KRAS)、肿瘤蛋白53(TP53)和B细胞淋巴瘤/白血病-2(BCL2)等关键基因。该技术在抑制肿瘤生长、修复突变和提高癌细胞对化疗的敏感性方面已显示出潜力。此外,CRISPR-Cas9 还被用于鉴定导致耐药性的新型关键基因和分子机制,为治疗干预提供了新途径。这篇综述还重点介绍了 CRISPR-Cas9 在针对免疫逃逸机制和开发提高药物敏感性策略方面的应用:CRISPR-Cas9技术在推进肺癌治疗,尤其是解决耐药性方面大有可为。精确靶向和编辑涉及耐药途径的基因的能力为开发更有效和个性化的疗法提供了强有力的工具。虽然在递送、安全性和伦理考虑方面仍存在挑战,但正在进行的研究和技术改进有望进一步加强 CRISPR-Cas9 在改善肺癌患者治疗效果方面的作用。
{"title":"Research into overcoming drug resistance in lung cancer treatment using CRISPR-Cas9 technology: a narrative review.","authors":"Bin Liu, Ziyu Wang, Meng Gu, Jinghui Wang, Jinjing Tan","doi":"10.21037/tlcr-24-592","DOIUrl":"https://doi.org/10.21037/tlcr-24-592","url":null,"abstract":"<p><strong>Background and objective: </strong>Lung cancer remains a leading cause of cancer-related mortality globally, with drug resistance posing a significant challenge to effective treatment. The advent of clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (CRISPR-Cas9) technology offers a novel and precise gene-editing technology for targeting and negating drug resistance mechanisms in lung cancer. This review summarizes the research progress in the use of CRISPR-Cas9 technology for investigating and managing drug resistance in lung cancer treatment.</p><p><strong>Methods: </strong>A literature search was conducted using the Web of Science and PubMed databases, with the following keywords: [CRISPR-Cas9], [lung cancer], [drug resistance], [gene editing], and [gene therapy]. The search was limited to articles published in English from 2002 to September 2023. From the search results, studies that utilized CRISPR-Cas9 technology in the context of lung cancer drug resistance were selected for further analysis and summarize.</p><p><strong>Key content and findings: </strong>CRISPR-Cas9 technology enables precise DNA-sequence editing, allowing for the targeted addition, deletion, or modification of genes. It has been applied to investigate drug resistance in lung cancer by focusing on key genes such as epidermal growth factor receptor (<i>EGFR</i>), Kirsten rat sarcoma viral oncogene homolog (<i>KRAS</i>), tumor protein 53 (<i>TP53</i>), and B-cell lymphoma/leukemia-2 (<i>BCL2</i>), among others. The technology has shown potential in inhibiting tumor growth, repairing mutations, and enhancing the sensitivity of cancer cells to chemotherapy. Additionally, CRISPR-Cas9 has been used to identify novel key genes and molecular mechanisms contributing to drug resistance, offering new avenues for therapeutic intervention. The review also highlights the use of CRISPR-Cas9 in targeting immune escape mechanisms and the development of strategies to improve drug sensitivity.</p><p><strong>Conclusions: </strong>The CRISPR-Cas9 technology holds great promise for advancing lung cancer treatment, particularly in addressing drug resistance. The ability to precisely target and edit genes involved in resistance pathways offers a powerful tool for developing more effective and personalized therapies. While challenges remain in terms of delivery, safety, and ethical considerations, ongoing research and technological refinements are expected to further enhance the role of CRISPR-Cas9 in improving patient outcomes in lung cancer treatment.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11384501/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142296344","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 (un)lucky seven-how can we mitigate risk factors for postoperative pneumonia after lung resections? 不幸的七人--如何降低肺切除术后肺炎的风险因素?
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-08-31 Epub Date: 2024-08-12 DOI: 10.21037/tlcr-24-428
Koen C H A Verkoulen, Iris E W G Laven, Jean H T Daemen, Juliette H R J Degens, Lizza E L Hendriks, Karel W E Hulsewé, Yvonne L J Vissers, Erik R de Loos
{"title":"The (un)lucky seven-how can we mitigate risk factors for postoperative pneumonia after lung resections?","authors":"Koen C H A Verkoulen, Iris E W G Laven, Jean H T Daemen, Juliette H R J Degens, Lizza E L Hendriks, Karel W E Hulsewé, Yvonne L J Vissers, Erik R de Loos","doi":"10.21037/tlcr-24-428","DOIUrl":"https://doi.org/10.21037/tlcr-24-428","url":null,"abstract":"","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11384492/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142296348","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
Efficacy and prognostic factors of stereotactic body radiotherapy combined with immunotherapy for pulmonary oligometastases: a preliminary retrospective cohort study. 立体定向体放射治疗联合免疫疗法治疗肺少转移灶的疗效和预后因素:一项初步回顾性队列研究。
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-08-31 Epub Date: 2024-08-28 DOI: 10.21037/tlcr-24-588
Mei-Na Piao, Jing Xie, Min-Min Jin, Xiao-Ting Ma, Zheng Dou, Jian-Ping Wang, Jin-Li Li

Background: Stereotactic body radiotherapy (SBRT) combined immunotherapy has a synergistic effect on patients with stage IV tumors. However, the efficacy and prognostic factors analysis of SBRT combined immunotherapy for patients with pulmonary oligometastases have rarely been reported in the studies. The purpose of this study is to explore the efficacy and prognostic factors analysis of SBRT combined immunotherapy for patients with oligometastatic lung tumors.

Methods: A retrospective analysis was conducted on 43 patients with advanced tumors who received SBRT combined with immunotherapy for pulmonary oligometastases from October 2018 to October 2021. Local control (LC), progression-free survival (PFS), and overall survival (OS) were assessed using the Kaplan-Meier method. Univariate and multivariate analyses of OS were performed using the Cox regression model, and the P value <0.05 was considered statistically significant. The receiver operating characteristic (ROC) curve of neutrophil-to-lymphocyte ratio (NLR) after SBRT was generated. Spearman correlation analysis was used to determine the relationship of planning target volume (PTV) with absolute lymphocyte count (ALC) before and after SBRT and with neutrophil count (NE) after SBRT. Additionally, linear regression was used to examine the relationship between ALC after SBRT and clinical factors.

Results: A total of 43 patients with pulmonary oligometastases receiving SBRT combined with immunotherapy were included in the study. The change in NLR after SBRT was statistically significant (P<0.001). At 1 and 2 years, respectively, the LC rates were 90.3% and 87.5%, the OS rates were 83.46% and 60.99%, and the PFS rates were 69.92% and 54.25%, with a median PFS of 27.00 (17.84-36.13) months. Univariate and multivariate Cox regression analyses showed that a shorter interval between radiotherapy and immunization [≤21 days; hazard ratio (HR) =1.10, 95% confidence interval (CI): 0.06-0.89; P=0.02] and a low NLR after SBRT (HR =0.24, 95% CI: 1.01-1.9; P=0.03) were associated with improved OS. The ROC curve identified 4.12 as the cutoff value for predicting OS based on NLR after SBRT. NLR after SBRT ≤4.12 significantly extended OS compared to NLR after SBRT >4.12 (log-rank P=0.001). Spearman correlation analysis and linear regression analysis showed that PTV was negatively correlated with ALC after SBRT.

Conclusions: Our preliminary research shows that SBRT combined with immunotherapy has a good effect, and NLR after SBRT is a poor prognostic factor for OS. Larger PTV volume is associated with decreased ALC after SBRT.

背景:立体定向体放射治疗(SBRT)联合免疫治疗对 IV 期肿瘤患者具有协同作用。然而,SBRT 联合免疫疗法对肺寡转移患者的疗效和预后因素分析却鲜有报道。本研究旨在探讨SBRT联合免疫疗法对少转移肺肿瘤患者的疗效和预后因素分析:对2018年10月至2021年10月接受SBRT联合免疫治疗的43例肺少转移瘤晚期肿瘤患者进行回顾性分析。采用Kaplan-Meier法评估了局部控制(LC)、无进展生存期(PFS)和总生存期(OS)。采用Cox回归模型对OS进行单变量和多变量分析,P值 结果:研究共纳入43例接受SBRT联合免疫疗法的肺寡转移患者。SBRT后NLR的变化具有统计学意义(P4.12(log-rank P=0.001))。Spearman相关性分析和线性回归分析表明,SBRT后PTV与ALC呈负相关:我们的初步研究表明,SBRT 联合免疫疗法具有良好的效果,而 SBRT 后的 NLR 是 OS 的不良预后因素。较大的PTV体积与SBRT后ALC的降低有关。
{"title":"Efficacy and prognostic factors of stereotactic body radiotherapy combined with immunotherapy for pulmonary oligometastases: a preliminary retrospective cohort study.","authors":"Mei-Na Piao, Jing Xie, Min-Min Jin, Xiao-Ting Ma, Zheng Dou, Jian-Ping Wang, Jin-Li Li","doi":"10.21037/tlcr-24-588","DOIUrl":"https://doi.org/10.21037/tlcr-24-588","url":null,"abstract":"<p><strong>Background: </strong>Stereotactic body radiotherapy (SBRT) combined immunotherapy has a synergistic effect on patients with stage IV tumors. However, the efficacy and prognostic factors analysis of SBRT combined immunotherapy for patients with pulmonary oligometastases have rarely been reported in the studies. The purpose of this study is to explore the efficacy and prognostic factors analysis of SBRT combined immunotherapy for patients with oligometastatic lung tumors.</p><p><strong>Methods: </strong>A retrospective analysis was conducted on 43 patients with advanced tumors who received SBRT combined with immunotherapy for pulmonary oligometastases from October 2018 to October 2021. Local control (LC), progression-free survival (PFS), and overall survival (OS) were assessed using the Kaplan-Meier method. Univariate and multivariate analyses of OS were performed using the Cox regression model, and the P value <0.05 was considered statistically significant. The receiver operating characteristic (ROC) curve of neutrophil-to-lymphocyte ratio (NLR) after SBRT was generated. Spearman correlation analysis was used to determine the relationship of planning target volume (PTV) with absolute lymphocyte count (ALC) before and after SBRT and with neutrophil count (NE) after SBRT. Additionally, linear regression was used to examine the relationship between ALC after SBRT and clinical factors.</p><p><strong>Results: </strong>A total of 43 patients with pulmonary oligometastases receiving SBRT combined with immunotherapy were included in the study. The change in NLR after SBRT was statistically significant (P<0.001). At 1 and 2 years, respectively, the LC rates were 90.3% and 87.5%, the OS rates were 83.46% and 60.99%, and the PFS rates were 69.92% and 54.25%, with a median PFS of 27.00 (17.84-36.13) months. Univariate and multivariate Cox regression analyses showed that a shorter interval between radiotherapy and immunization [≤21 days; hazard ratio (HR) =1.10, 95% confidence interval (CI): 0.06-0.89; P=0.02] and a low NLR after SBRT (HR =0.24, 95% CI: 1.01-1.9; P=0.03) were associated with improved OS. The ROC curve identified 4.12 as the cutoff value for predicting OS based on NLR after SBRT. NLR after SBRT ≤4.12 significantly extended OS compared to NLR after SBRT >4.12 (log-rank P=0.001). Spearman correlation analysis and linear regression analysis showed that PTV was negatively correlated with ALC after SBRT.</p><p><strong>Conclusions: </strong>Our preliminary research shows that SBRT combined with immunotherapy has a good effect, and NLR after SBRT is a poor prognostic factor for OS. Larger PTV volume is associated with decreased ALC after SBRT.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11384503/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142296329","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
Clinical insights: five-year follow-up of KEYNOTE-189 trial outcomes and more. 临床见解:KEYNOTE-189 试验结果及更多信息的五年随访。
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-08-31 Epub Date: 2024-08-05 DOI: 10.21037/tlcr-24-198
Bobby Se, Athar Eysa, Nagla Karim
{"title":"Clinical insights: five-year follow-up of KEYNOTE-189 trial outcomes and more.","authors":"Bobby Se, Athar Eysa, Nagla Karim","doi":"10.21037/tlcr-24-198","DOIUrl":"https://doi.org/10.21037/tlcr-24-198","url":null,"abstract":"","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11384474/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142296324","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 nomogram predicting the risk of extrathoracic metastasis at initial diagnosis of T≤3cmN0 lung cancer. 预测 T≤3cmN0 肺癌初诊时胸外转移风险的提名图。
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-08-31 Epub Date: 2024-08-08 DOI: 10.21037/tlcr-24-338
Tengyong Wang, Zihuai Wang, Jian Zhou, Hui Jie, Hu Liao, Jiandong Mei, Qiang Pu, Lunxu Liu

Background: The risk and risk factors of extrathoracic metastasis at initial diagnosis in T≤3cmN0 lung cancer patients are not fully understood. We aimed to develop a model to predict the risk of extrathoracic metastasis in those patients.

Methods: Clinicopathological data of patients were collected from Surveillance, Epidemiology, and End Results (SEER) database. Univariable and multivariable analyses using logistic regression were conducted to identify risk factors. A predictive model and corresponding nomogram were developed based on the risk factors. The model was assessed using the area under the receiver operating characteristic curve (AUC), Hosmer-Lemeshow test, and decision curve.

Results: A total of 20,057 T≤3cmN0 patients were enrolled, of whom 251 (1.25%) were diagnosed with extrathoracic metastasis at the initial diagnosis. Aged ≤50 [odds ratio (OR): 2.05, 95% confidence interval (CI): 1.19-3.53, P=0.01] and aged ≥81 [1.65 (1.05-2.58), P=0.03], Hispanic [1.81 (1.20-2.71), P=0.004], location of bronchus [3.18 (1.08-9.35), P=0.04], larger tumor size, pleural invasion, and a history of colorectal cancer [2.01 (1.01-4.00), P=0.046] were independent risk factors. In the training cohort and validation cohort, the AUCs of the developed model were 0.727, 0.728 respectively, and the results of Hosmer-Lemeshow test were P=0.47, P=0.61 respectively. The decision curve showed good clinical meaning of the model.

Conclusions: Extrathoracic metastasis at initial diagnosis in T≤3cmN0 lung cancer patients was not rare. The model based on the risk factors showed good performance in predicting the risk of extrathoracic metastasis.

背景:T≤3cmN0肺癌患者在初诊时发生胸外转移的风险和风险因素尚不完全清楚。我们旨在建立一个模型来预测这些患者发生胸外转移的风险:方法:从监测、流行病学和最终结果(SEER)数据库中收集患者的临床病理数据。采用逻辑回归法进行单变量和多变量分析,以确定风险因素。根据风险因素建立了预测模型和相应的提名图。使用接收者操作特征曲线下面积(AUC)、Hosmer-Lemeshow 检验和决策曲线对模型进行评估:共有20,057例T≤3cmN0患者入选,其中251例(1.25%)在初诊时被诊断为胸外转移。年龄≤50岁[几率比(OR):2.05,95%置信区间(CI):1.19-3.53,P=0.01]和年龄≥81岁[1.65(1.05-2.58),P=0.03]、西班牙裔[1.81(1.20-2.71),P=0.004]、支气管位置[3.18 (1.08-9.35),P=0.04]、肿瘤较大、胸膜侵犯和结直肠癌病史[2.01 (1.01-4.00),P=0.046]是独立的风险因素。在训练队列和验证队列中,所建立模型的AUC分别为0.727和0.728,Hosmer-Lemeshow检验结果分别为P=0.47和P=0.61。决策曲线显示该模型具有良好的临床意义:结论:T≤3cmN0肺癌患者初诊时发生胸外转移并不罕见。基于风险因素的模型在预测胸外转移风险方面表现良好。
{"title":"A nomogram predicting the risk of extrathoracic metastasis at initial diagnosis of T<sub>≤3cm</sub>N<sub>0</sub> lung cancer.","authors":"Tengyong Wang, Zihuai Wang, Jian Zhou, Hui Jie, Hu Liao, Jiandong Mei, Qiang Pu, Lunxu Liu","doi":"10.21037/tlcr-24-338","DOIUrl":"https://doi.org/10.21037/tlcr-24-338","url":null,"abstract":"<p><strong>Background: </strong>The risk and risk factors of extrathoracic metastasis at initial diagnosis in T<sub>≤3cm</sub>N<sub>0</sub> lung cancer patients are not fully understood. We aimed to develop a model to predict the risk of extrathoracic metastasis in those patients.</p><p><strong>Methods: </strong>Clinicopathological data of patients were collected from Surveillance, Epidemiology, and End Results (SEER) database. Univariable and multivariable analyses using logistic regression were conducted to identify risk factors. A predictive model and corresponding nomogram were developed based on the risk factors. The model was assessed using the area under the receiver operating characteristic curve (AUC), Hosmer-Lemeshow test, and decision curve.</p><p><strong>Results: </strong>A total of 20,057 T<sub>≤3cm</sub>N<sub>0</sub> patients were enrolled, of whom 251 (1.25%) were diagnosed with extrathoracic metastasis at the initial diagnosis. Aged ≤50 [odds ratio (OR): 2.05, 95% confidence interval (CI): 1.19-3.53, P=0.01] and aged ≥81 [1.65 (1.05-2.58), P=0.03], Hispanic [1.81 (1.20-2.71), P=0.004], location of bronchus [3.18 (1.08-9.35), P=0.04], larger tumor size, pleural invasion, and a history of colorectal cancer [2.01 (1.01-4.00), P=0.046] were independent risk factors. In the training cohort and validation cohort, the AUCs of the developed model were 0.727, 0.728 respectively, and the results of Hosmer-Lemeshow test were P=0.47, P=0.61 respectively. The decision curve showed good clinical meaning of the model.</p><p><strong>Conclusions: </strong>Extrathoracic metastasis at initial diagnosis in T<sub>≤3cm</sub>N<sub>0</sub> lung cancer patients was not rare. The model based on the risk factors showed good performance in predicting the risk of extrathoracic metastasis.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11384498/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142296318","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
REGN5093-M114: can an antibody-drug conjugate overcome the challenge of resistance to epidermal growth factor receptor and mesenchymal epithelial transition tyrosine kinase inhibitors in non-small cell lung cancer? REGN5093-M114:抗体药物共轭物能否克服非小细胞肺癌患者对表皮生长因子受体和间质上皮转化酪氨酸激酶抑制剂的耐药性挑战?
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-08-31 Epub Date: 2024-07-26 DOI: 10.21037/tlcr-24-144
Julie Dardare, Andréa Witz, Alexandre Harlé
{"title":"REGN5093-M114: can an antibody-drug conjugate overcome the challenge of resistance to epidermal growth factor receptor and mesenchymal epithelial transition tyrosine kinase inhibitors in non-small cell lung cancer?","authors":"Julie Dardare, Andréa Witz, Alexandre Harlé","doi":"10.21037/tlcr-24-144","DOIUrl":"https://doi.org/10.21037/tlcr-24-144","url":null,"abstract":"","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11384497/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142296343","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
LungPath: artificial intelligence-driven histologic pattern recognition for improved diagnosis of early-stage invasive lung adenocarcinoma. LungPath:人工智能驱动的组织学模式识别,用于改进早期浸润性肺腺癌的诊断。
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-08-31 Epub Date: 2024-08-26 DOI: 10.21037/tlcr-24-258
Haoda Huang, Zeping Yan, Bingliang Li, Weixiang Lu, Ping He, Lei Fan, Xiaowei Wu, Hengrui Liang, Jianxing He

Background: Early-stage invasive lung adenocarcinoma (ADC) characterized by a predominant micropapillary or solid pattern exhibit an elevated risk of recurrence following sub-lobar resection, thus determining histological subtype of early-stage invasive ADC prior surgery is important for formulating lobectomy or sub-lobar resection. This study aims to develop a deep learning algorithm and assess its clinical capability in distinguishing high-risk or low-risk histologic patterns in early-stage invasive ADC based on preoperative computed tomography (CT) scans.

Methods: Two retrospective cohorts were included: development cohort 1 and external test cohort 2, comprising patients diagnosed with T1 stage invasive ADC. Electronic medical records and CT scans of all patients were documented. Patients were stratified into two risk groups. High-risk group: comprising cases with a micropapillary component ≥5% or a predominant solid pattern. Low-risk group: encompassing cases with a micropapillary component <5% and an absence of a predominant solid pattern. The overall segmentation model was modified based on Mask Region-based Convolutional Neural Network (Mask-RCNN), and Residual Network 50 (ResNet50)_3D was employed for image classification.

Results: A total of 432 patients participated in this study, with 385 cases in cohort 1 and 47 cases in cohort 2. The fine-outline results produced by the auto-segmentation model exhibited a high level of agreement with manual segmentation by human experts, yielding a mean dice coefficient of 0.86 [95% confidence interval (CI): 0.85-0.87] in cohort 1 and 0.84 (95% CI: 0.82-0.85) in cohort 2. Furthermore, the deep learning model effectively differentiated the high-risk group from the low-risk group, achieving an area under the curve (AUC) of 0.89 (95% CI: 0.88-0.90) in cohort 1. In the external validation conducted in cohort 2, the deep learning model displayed an AUC of 0.87 (95% CI: 0.84-0.88) in distinguishing the high-risk group from the low-risk group. The average diagnostic time was 16.00±3.2 seconds, with an accuracy of 0.82 (95% CI: 0.81-0.83).

Conclusions: We have developed a deep learning algorithm, LungPath, for the automated segmentation of pulmonary nodules and prediction of high-risk histological patterns in early-stage lung ADC based on CT scans.

背景:早期浸润性肺腺癌(ADC)以微乳头状或实性形态为主,在肺叶下切除术后复发风险较高,因此术前确定早期浸润性ADC的组织学亚型对于制定肺叶切除术或肺叶下切除术非常重要。本研究旨在开发一种深度学习算法,并评估其根据术前计算机断层扫描(CT)区分早期浸润性ADC的高风险或低风险组织学模式的临床能力:纳入两个回顾性队列:开发队列 1 和外部测试队列 2,包括确诊为 T1 期浸润性 ADC 的患者。记录了所有患者的电子病历和 CT 扫描结果。患者被分为两个风险组。高风险组:包括微乳头成分≥5%或以实性形态为主的病例。低风险组:包括微乳头成分的病例 结果:共有 432 名患者参与了这项研究,其中第一组 385 例,第二组 47 例。自动分割模型生成的精细外线结果与人类专家的人工分割结果具有很高的一致性,组群 1 的平均骰子系数为 0.86 [95% 置信区间 (CI):0.85-0.87],组群 2 的平均骰子系数为 0.84 (95% CI:0.82-0.85)。此外,深度学习模型有效区分了高风险组和低风险组,在队列 1 中的曲线下面积(AUC)达到了 0.89(95% CI:0.88-0.90)。在队列 2 中进行的外部验证中,深度学习模型在区分高风险组和低风险组方面的 AUC 为 0.87(95% CI:0.84-0.88)。平均诊断时间为(16.00±3.2)秒,准确率为 0.82(95% CI:0.81-0.83):我们开发了一种深度学习算法 LungPath,用于基于 CT 扫描自动分割肺结节和预测早期肺 ADC 的高危组织学模式。
{"title":"<i>LungPath</i>: artificial intelligence-driven histologic pattern recognition for improved diagnosis of early-stage invasive lung adenocarcinoma.","authors":"Haoda Huang, Zeping Yan, Bingliang Li, Weixiang Lu, Ping He, Lei Fan, Xiaowei Wu, Hengrui Liang, Jianxing He","doi":"10.21037/tlcr-24-258","DOIUrl":"https://doi.org/10.21037/tlcr-24-258","url":null,"abstract":"<p><strong>Background: </strong>Early-stage invasive lung adenocarcinoma (ADC) characterized by a predominant micropapillary or solid pattern exhibit an elevated risk of recurrence following sub-lobar resection, thus determining histological subtype of early-stage invasive ADC prior surgery is important for formulating lobectomy or sub-lobar resection. This study aims to develop a deep learning algorithm and assess its clinical capability in distinguishing high-risk or low-risk histologic patterns in early-stage invasive ADC based on preoperative computed tomography (CT) scans.</p><p><strong>Methods: </strong>Two retrospective cohorts were included: development cohort 1 and external test cohort 2, comprising patients diagnosed with T1 stage invasive ADC. Electronic medical records and CT scans of all patients were documented. Patients were stratified into two risk groups. High-risk group: comprising cases with a micropapillary component ≥5% or a predominant solid pattern. Low-risk group: encompassing cases with a micropapillary component <5% and an absence of a predominant solid pattern. The overall segmentation model was modified based on Mask Region-based Convolutional Neural Network (Mask-RCNN), and Residual Network 50 (ResNet50)_3D was employed for image classification.</p><p><strong>Results: </strong>A total of 432 patients participated in this study, with 385 cases in cohort 1 and 47 cases in cohort 2. The fine-outline results produced by the auto-segmentation model exhibited a high level of agreement with manual segmentation by human experts, yielding a mean dice coefficient of 0.86 [95% confidence interval (CI): 0.85-0.87] in cohort 1 and 0.84 (95% CI: 0.82-0.85) in cohort 2. Furthermore, the deep learning model effectively differentiated the high-risk group from the low-risk group, achieving an area under the curve (AUC) of 0.89 (95% CI: 0.88-0.90) in cohort 1. In the external validation conducted in cohort 2, the deep learning model displayed an AUC of 0.87 (95% CI: 0.84-0.88) in distinguishing the high-risk group from the low-risk group. The average diagnostic time was 16.00±3.2 seconds, with an accuracy of 0.82 (95% CI: 0.81-0.83).</p><p><strong>Conclusions: </strong>We have developed a deep learning algorithm, <i>LungPath</i>, for the automated segmentation of pulmonary nodules and prediction of high-risk histological patterns in early-stage lung ADC based on CT scans.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11384487/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142296317","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
Oncogene-addicted solid tumors and microbiome-lung cancer as a main character: a narrative review. 肿瘤基因上瘾的实体瘤和作为主角的微生物组--肺癌:叙述性综述。
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-08-31 Epub Date: 2024-08-06 DOI: 10.21037/tlcr-24-216
Mora Guardamagna, May-Lucie Meyer, Miguel Ángel Berciano-Guerrero, Andres Mesas-Ruiz, Manuel Cobo-Dols, Elisabeth Perez-Ruiz, Alexandra Cantero Gonzalez, Rocío Lavado-Valenzuela, Isabel Barragán, Javier Oliver, Alicia Garrido-Aranda, Martina Alvarez, Antonio Rueda-Dominguez, María Isabel Queipo-Ortuño, Emilio Alba Conejo, Jose Carlos Benitez

Background and objective: Lung cancer stands as the main cause of cancer-related deaths worldwide. With the advent of immunotherapy and the discovery of targetable oncogenic driver genes, although prognosis has changed in the last few years, survival rates remain dismal for most patients. This emphasizes the urgent need for new strategies that could enhance treatment in precision medicine. The role of the microbiota in carcinogenesis constitutes an evolving landscape of which little is known. It has been suggested these microorganisms may influence in responses, resistance, and adverse effects to cancer treatments, particularly to immune checkpoint blockers. However, evidence on the impact of microbiota composition in oncogene-addicted tumors is lacking. This review aims to provide an overview of the relationship between microbiota, daily habits, the immune system, and oncogene-addicted tumors, focusing on lung cancer.

Methods: A PubMed and Google Scholar search from 2013 to 2024 was conducted. Relevant articles were reviewed in order to guide our research and generate hypothesis of clinical applicability.

Key content and findings: Microbiota is recognized to participate in immune reprogramming, fostering inflammatory, immunosuppressive, or anti-tumor responses. Therefore, identifying the microbiota that impact response to treatment and modulating its composition by interventions such as dietary modifications, probiotics or antibiotics, could potentially yield better outcomes for cancer patients. Additionally, targeted therapies that modulate molecular signaling pathways may impact both immunity and microbiota. Understanding this intricate interplay could unveil new therapeutic strategies.

Conclusions: By comprehending how microbiota may influence efficacy of targeted therapies, even though current evidence is scarce, we may generate interesting hypotheses that could improve clinical practice.

背景和目的:肺癌是全球癌症相关死亡的主要原因。随着免疫疗法的出现和可靶向致癌驱动基因的发现,虽然预后在过去几年中有所改变,但大多数患者的生存率仍然不容乐观。这凸显了人们对新策略的迫切需求,新策略可以提高精准医疗的治疗效果。微生物群在致癌过程中的作用是一个不断演变的过程,目前人们对此知之甚少。有人认为,这些微生物可能会影响癌症治疗的反应、抗药性和不良反应,尤其是对免疫检查点阻断剂的影响。然而,关于微生物群组成对使用癌基因的肿瘤的影响还缺乏证据。本综述旨在概述微生物群、日常生活习惯、免疫系统和致癌基因肿瘤之间的关系,重点关注肺癌:方法:对2013年至2024年的PubMed和Google Scholar进行了搜索。对相关文章进行了综述,以指导我们的研究并提出临床适用性假设:微生物群被认为可参与免疫重编程,促进炎症、免疫抑制或抗肿瘤反应。因此,确定影响治疗反应的微生物群,并通过饮食调整、益生菌或抗生素等干预措施调节其组成,有可能为癌症患者带来更好的治疗效果。此外,调节分子信号通路的靶向疗法可能会对免疫和微生物群产生影响。了解这种错综复杂的相互作用可以揭示新的治疗策略:通过了解微生物群如何影响靶向疗法的疗效(尽管目前的证据还很少),我们可能会提出一些有趣的假设,从而改善临床实践。
{"title":"Oncogene-addicted solid tumors and microbiome-lung cancer as a main character: a narrative review.","authors":"Mora Guardamagna, May-Lucie Meyer, Miguel Ángel Berciano-Guerrero, Andres Mesas-Ruiz, Manuel Cobo-Dols, Elisabeth Perez-Ruiz, Alexandra Cantero Gonzalez, Rocío Lavado-Valenzuela, Isabel Barragán, Javier Oliver, Alicia Garrido-Aranda, Martina Alvarez, Antonio Rueda-Dominguez, María Isabel Queipo-Ortuño, Emilio Alba Conejo, Jose Carlos Benitez","doi":"10.21037/tlcr-24-216","DOIUrl":"https://doi.org/10.21037/tlcr-24-216","url":null,"abstract":"<p><strong>Background and objective: </strong>Lung cancer stands as the main cause of cancer-related deaths worldwide. With the advent of immunotherapy and the discovery of targetable oncogenic driver genes, although prognosis has changed in the last few years, survival rates remain dismal for most patients. This emphasizes the urgent need for new strategies that could enhance treatment in precision medicine. The role of the microbiota in carcinogenesis constitutes an evolving landscape of which little is known. It has been suggested these microorganisms may influence in responses, resistance, and adverse effects to cancer treatments, particularly to immune checkpoint blockers. However, evidence on the impact of microbiota composition in oncogene-addicted tumors is lacking. This review aims to provide an overview of the relationship between microbiota, daily habits, the immune system, and oncogene-addicted tumors, focusing on lung cancer.</p><p><strong>Methods: </strong>A PubMed and Google Scholar search from 2013 to 2024 was conducted. Relevant articles were reviewed in order to guide our research and generate hypothesis of clinical applicability.</p><p><strong>Key content and findings: </strong>Microbiota is recognized to participate in immune reprogramming, fostering inflammatory, immunosuppressive, or anti-tumor responses. Therefore, identifying the microbiota that impact response to treatment and modulating its composition by interventions such as dietary modifications, probiotics or antibiotics, could potentially yield better outcomes for cancer patients. Additionally, targeted therapies that modulate molecular signaling pathways may impact both immunity and microbiota. Understanding this intricate interplay could unveil new therapeutic strategies.</p><p><strong>Conclusions: </strong>By comprehending how microbiota may influence efficacy of targeted therapies, even though current evidence is scarce, we may generate interesting hypotheses that could improve clinical practice.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11384476/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142296338","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
Neutrophil estimation and prognosis analysis based on existing lung squamous cell carcinoma datasets: the development and validation of a prognosis prediction model. 基于现有肺鳞状细胞癌数据集的中性粒细胞估计和预后分析:预后预测模型的开发与验证。
IF 4 2区 医学 Q2 ONCOLOGY Pub Date : 2024-08-31 Epub Date: 2024-08-19 DOI: 10.21037/tlcr-24-411
Youyu Wang, Dongfang Li, Qiang Li, Alina Basnet, Jimmy T Efird, Nobuhiko Seki
<p><strong>Background: </strong>Notwithstanding the rapid developments in precision medicine in recent years, lung cancer still has a low survival rate, especially lung squamous cell cancer (LUSC). The tumor microenvironment (TME) plays an important role in the progression of lung cancer, in which high neutrophil levels are correlated with poor prognosis, potentially due to their interactions with tumor cells via pro-inflammatory cytokines and chemokines. However, the precise mechanisms of how neutrophils influence lung cancer remain unclear. This study aims to explore these mechanisms and develop a prognosis predictive model in LUSC, addressing the knowledge gap in neutrophil-related cancer pathogenesis.</p><p><strong>Methods: </strong>LUSC datasets from the Xena Hub and Gene Expression Omnibus (GEO) databases were used, comprising 473 tumor samples and 195 tumor samples, respectively. Neutrophil contents in these samples were estimated using CIBERSORT, xCell, and microenvironment cell populations (MCP) counter tools. Differentially expressed genes (DEGs) were identified using DEseq2, and a weighted gene co-expression network analysis (WGCNA) was performed to identify neutrophil-related genes. A least absolute shrinkage and selection operator (LASSO) Cox regression model was constructed for prognosis prediction, and the model's accuracy was validated using Kaplan-Meier survival curves and time-dependent receiver operating characteristic (ROC) curves. Additionally, genomic changes, immune correlations, drug sensitivity, and immunotherapy response were analyzed to further validate the model's predictive power.</p><p><strong>Results: </strong>Neutrophil content was significantly higher in adjacent normal tissue compared to LUSC tissue (P<0.001). High neutrophil content was associated with worse overall survival (OS) (P=0.02), disease-free survival (DFS) (P=0.02), and progression-free survival (PFS) (P=0.03) using different software estimates. Nine gene modules were identified, with blue and yellow modules showing strong correlations with neutrophil prognosis (P<0.001). Eight genes were selected for the prognostic model, which accurately predicted 1-, 3-, and 5-year survival in both the training set [area under the curve (AUC) value =0.60, 0.63, 0.66, respectively] and validation set (AUC value =0.58, 0.58, 0.59, respectively), with significant prognosis differences between high- and low-risk groups (P<0.001). The model's independent prognostic factors included risk group, pathologic M stage, and tumor stage (P<0.05). A further molecular mechanism analysis revealed differences between risk groups were revealed in immune checkpoint and human leukocyte antigen (HLA) gene expression, hallmark pathways, drug sensitivity, and immunotherapy responses.</p><p><strong>Conclusions: </strong>This study established a risk-score model that effectively predicts the prognosis of LUSC patients and sheds light on the molecular mechanisms involved. The findings enhan
背景:尽管近年来精准医疗发展迅速,但肺癌的生存率仍然很低,尤其是肺鳞癌(LUSC)。肿瘤微环境(TME)在肺癌的进展中起着重要作用,其中中性粒细胞水平高与预后不良相关,这可能是由于中性粒细胞通过促炎细胞因子和趋化因子与肿瘤细胞相互作用所致。然而,中性粒细胞影响肺癌的确切机制仍不清楚。本研究旨在探索这些机制,并建立一个肺癌预后预测模型,填补中性粒细胞相关癌症发病机制方面的知识空白:研究使用了来自 Xena Hub 和 Gene Expression Omnibus (GEO) 数据库的 LUSC 数据集,分别包括 473 个肿瘤样本和 195 个肿瘤样本。利用CIBERSORT、xCell和微环境细胞群(MCP)计数工具估算了这些样本中的中性粒细胞含量。使用 DEseq2 鉴定了差异表达基因(DEGs),并进行了加权基因共表达网络分析(WGCNA)以鉴定中性粒细胞相关基因。构建了最小绝对收缩和选择算子(LASSO)Cox回归模型用于预后预测,并利用Kaplan-Meier生存曲线和随时间变化的接收者操作特征曲线(ROC)验证了该模型的准确性。此外,还对基因组变化、免疫相关性、药物敏感性和免疫治疗反应进行了分析,以进一步验证模型的预测能力:结果:与 LUSC 组织相比,邻近正常组织的中性粒细胞含量明显更高(PConclusions:本研究建立的风险评分模型可有效预测LUSC患者的预后,并揭示了其中的分子机制。研究结果加深了人们对中性粒细胞与肿瘤相互作用的理解,为个性化治疗提供了潜在靶点。然而,还需要进一步的实验验证和临床研究来证实这些发现,并解决研究的局限性,包括对公共数据库的依赖和对特定肺癌亚型的关注。
{"title":"Neutrophil estimation and prognosis analysis based on existing lung squamous cell carcinoma datasets: the development and validation of a prognosis prediction model.","authors":"Youyu Wang, Dongfang Li, Qiang Li, Alina Basnet, Jimmy T Efird, Nobuhiko Seki","doi":"10.21037/tlcr-24-411","DOIUrl":"https://doi.org/10.21037/tlcr-24-411","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;Notwithstanding the rapid developments in precision medicine in recent years, lung cancer still has a low survival rate, especially lung squamous cell cancer (LUSC). The tumor microenvironment (TME) plays an important role in the progression of lung cancer, in which high neutrophil levels are correlated with poor prognosis, potentially due to their interactions with tumor cells via pro-inflammatory cytokines and chemokines. However, the precise mechanisms of how neutrophils influence lung cancer remain unclear. This study aims to explore these mechanisms and develop a prognosis predictive model in LUSC, addressing the knowledge gap in neutrophil-related cancer pathogenesis.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;LUSC datasets from the Xena Hub and Gene Expression Omnibus (GEO) databases were used, comprising 473 tumor samples and 195 tumor samples, respectively. Neutrophil contents in these samples were estimated using CIBERSORT, xCell, and microenvironment cell populations (MCP) counter tools. Differentially expressed genes (DEGs) were identified using DEseq2, and a weighted gene co-expression network analysis (WGCNA) was performed to identify neutrophil-related genes. A least absolute shrinkage and selection operator (LASSO) Cox regression model was constructed for prognosis prediction, and the model's accuracy was validated using Kaplan-Meier survival curves and time-dependent receiver operating characteristic (ROC) curves. Additionally, genomic changes, immune correlations, drug sensitivity, and immunotherapy response were analyzed to further validate the model's predictive power.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;Neutrophil content was significantly higher in adjacent normal tissue compared to LUSC tissue (P&lt;0.001). High neutrophil content was associated with worse overall survival (OS) (P=0.02), disease-free survival (DFS) (P=0.02), and progression-free survival (PFS) (P=0.03) using different software estimates. Nine gene modules were identified, with blue and yellow modules showing strong correlations with neutrophil prognosis (P&lt;0.001). Eight genes were selected for the prognostic model, which accurately predicted 1-, 3-, and 5-year survival in both the training set [area under the curve (AUC) value =0.60, 0.63, 0.66, respectively] and validation set (AUC value =0.58, 0.58, 0.59, respectively), with significant prognosis differences between high- and low-risk groups (P&lt;0.001). The model's independent prognostic factors included risk group, pathologic M stage, and tumor stage (P&lt;0.05). A further molecular mechanism analysis revealed differences between risk groups were revealed in immune checkpoint and human leukocyte antigen (HLA) gene expression, hallmark pathways, drug sensitivity, and immunotherapy responses.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Conclusions: &lt;/strong&gt;This study established a risk-score model that effectively predicts the prognosis of LUSC patients and sheds light on the molecular mechanisms involved. The findings enhan","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-08-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11384491/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142296337","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