Background: Based on current technology, the accuracy of detecting malignancy in solitary pulmonary nodules (SPNs) is limited. This study aimed to establish a malignant risk prediction model for SPNs 5-15 mm in diameter.
Methods: We collected clinical characteristics and imaging features from 317 patients with SPNs 5-15 mm in diameter from the 900th Hospital of the Joint Logistic Support Force as a training cohort and 100 patients with SPNs 5-15 mm in diameter as a validation cohort. Univariate logistic regression analysis, least absolute shrinkage and selection operator (LASSO), and binary logistic regression analysis were used to screen for the independent influencing factors of benign and malignant SPN and to establish a prediction model for benign and malignant SPN with a diameter of 5-15 mm. The model in this study was compared with the Mayo model, Veterans Affairs (VA) model, Brock model, and Peking University People's Hospital (PKUPH) model. Finally, the clinical application value of this model was assessed.
Results: Univariate logistic regression analysis showed that smoking history, nodule diameter, nodule location, nodule density, margin, calcification, lobulation sign, spiculation sign, and vascular cluster sign were statistically significant factors. The results of LASSO and binary logistic regression analysis showed that smoking history, nodule diameter, nodule density, margin, lobulation sign, and vascular cluster sign were independent influencing factors of SPNs. The prediction model was successfully constructed and demonstrated a good predictive performance, with an area under the curve (AUC) value of 0.814 [95% confidence interval (CI): 0.768-0.861; P<0.001] in the training cohort and 0.864 (95% CI: 0.794-0.934; P<0.001) in the validation cohort. This model was shown to be highly accurate in predicting malignant SPNs and thus has a high clinical application value. Compared with previously described prediction models, including the Mayo model, VA model, Brock model, and PKUPH model, the proposed model demonstrated a significantly superior predictive ability.
Conclusions: The prediction model developed in this study can be used as an early screening method for SPNs 5-15 mm in diameter.
{"title":"Construction of a risk prediction model for isolated pulmonary nodules 5-15 mm in diameter.","authors":"Siting Xie, Xingguang Luo, Yuxin Guo, Xiulian Huang, Jinyu Long, Ying Chen, Ping Lin, Jinhe Xu, Shangwen Xu, Chunlei Zhao, Baoquan Lin, Chunxia Su, Nagarashee Seetharamu, Duilio Divisi, Mingliang Jin, Zongyang Yu","doi":"10.21037/tlcr-24-785","DOIUrl":"10.21037/tlcr-24-785","url":null,"abstract":"<p><strong>Background: </strong>Based on current technology, the accuracy of detecting malignancy in solitary pulmonary nodules (SPNs) is limited. This study aimed to establish a malignant risk prediction model for SPNs 5-15 mm in diameter.</p><p><strong>Methods: </strong>We collected clinical characteristics and imaging features from 317 patients with SPNs 5-15 mm in diameter from the 900th Hospital of the Joint Logistic Support Force as a training cohort and 100 patients with SPNs 5-15 mm in diameter as a validation cohort. Univariate logistic regression analysis, least absolute shrinkage and selection operator (LASSO), and binary logistic regression analysis were used to screen for the independent influencing factors of benign and malignant SPN and to establish a prediction model for benign and malignant SPN with a diameter of 5-15 mm. The model in this study was compared with the Mayo model, Veterans Affairs (VA) model, Brock model, and Peking University People's Hospital (PKUPH) model. Finally, the clinical application value of this model was assessed.</p><p><strong>Results: </strong>Univariate logistic regression analysis showed that smoking history, nodule diameter, nodule location, nodule density, margin, calcification, lobulation sign, spiculation sign, and vascular cluster sign were statistically significant factors. The results of LASSO and binary logistic regression analysis showed that smoking history, nodule diameter, nodule density, margin, lobulation sign, and vascular cluster sign were independent influencing factors of SPNs. The prediction model was successfully constructed and demonstrated a good predictive performance, with an area under the curve (AUC) value of 0.814 [95% confidence interval (CI): 0.768-0.861; P<0.001] in the training cohort and 0.864 (95% CI: 0.794-0.934; P<0.001) in the validation cohort. This model was shown to be highly accurate in predicting malignant SPNs and thus has a high clinical application value. Compared with previously described prediction models, including the Mayo model, VA model, Brock model, and PKUPH model, the proposed model demonstrated a significantly superior predictive ability.</p><p><strong>Conclusions: </strong>The prediction model developed in this study can be used as an early screening method for SPNs 5-15 mm in diameter.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 11","pages":"3139-3151"},"PeriodicalIF":4.0,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11632445/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142819011","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}
Pub Date : 2024-11-30Epub Date: 2024-11-28DOI: 10.21037/tlcr-24-525
Sarah E Herlihy, Caren Gentile, Samantha J Scott, Brandon A Smith, Kathryn A Stoll, Kala F Schilter, Justyna M Mordaka, Rebecca N Palmer, Christina Xyrafaki, Elizabeth Gillon-Zhang, Candace King, Ryan T Evans, Amanda S Green, Ana-Luisa Silva, Magdalena Stolarek-Januszkiewicz, Kristine von Bargen, Iyelola Turner, Chau Ha Ho, Alejandra Collazos, Nicola D Potts, Dilyara Nugent, Jinsy Jose, Eleanor R Gray, Elyse Shapiro, Wendy J Levin, Aishling Cooke, Barnaby W Balmforth, Robert J Osborne, Honey V Reddi, Vivianna M Van Deerlin
Background: Many patients with non-small cell lung cancer (NSCLC) lack access to highly effective approved targeted therapeutics due to multiple gaps in biomarker testing. Challenges in comprehensive molecular testing include complexities associated with the need to assess the presence of multiple variants, costs of running multiple sequential assays per sample, high assay quality control (QC) failure rates, clinical need for rapid turn-around time (TAT) to initiate therapy, and insufficient tissue samples. The ASPYRE-Lung NSCLC assay addresses gaps in multiplexed testing by simultaneously analyzing DNA and RNA, detecting 114 actionable genomic variants across 11 genes, consistent with current NSCLC treatment guidelines. This study was to assess the ease of adoption and performance of ASPYRE-Lung in third-party laboratories, comparing concordance across sites and with orthogonal methods.
Methods: ASPYRE-Lung was established at two academic centers with multiple operators per site. Assay concordance was evaluated across three sites using 77 patient samples [61 derived from formalin-fixed paraffin-embedded (FFPE) tissue and 16 from cytology specimens].
Results: Reproducibility for all 77 samples yielded a positive percent agreement (PPA) of 100% and negative percent agreement (NPA) of 99.99%. Concordance with next-generation sequencing (NGS)-based methods across all three sites was high with PPA of 97.2% and NPA of 99.96%.
Conclusions: ASPYRE-Lung assay is a cost-effective, easy to adopt testing method requiring no specialized expertise or complicated bioinformatics, with the potential to inform genomic data on small tissue samples, thus enabling all patients with NSCLC to undergo biomarker testing in a timely manner and benefit from appropriate targeted therapies.
{"title":"Evaluation of the ASPYRE-Lung targeted variant panel: a rapid, low-input solution for non-small cell lung cancer biomarker testing and experience from three independent sites.","authors":"Sarah E Herlihy, Caren Gentile, Samantha J Scott, Brandon A Smith, Kathryn A Stoll, Kala F Schilter, Justyna M Mordaka, Rebecca N Palmer, Christina Xyrafaki, Elizabeth Gillon-Zhang, Candace King, Ryan T Evans, Amanda S Green, Ana-Luisa Silva, Magdalena Stolarek-Januszkiewicz, Kristine von Bargen, Iyelola Turner, Chau Ha Ho, Alejandra Collazos, Nicola D Potts, Dilyara Nugent, Jinsy Jose, Eleanor R Gray, Elyse Shapiro, Wendy J Levin, Aishling Cooke, Barnaby W Balmforth, Robert J Osborne, Honey V Reddi, Vivianna M Van Deerlin","doi":"10.21037/tlcr-24-525","DOIUrl":"10.21037/tlcr-24-525","url":null,"abstract":"<p><strong>Background: </strong>Many patients with non-small cell lung cancer (NSCLC) lack access to highly effective approved targeted therapeutics due to multiple gaps in biomarker testing. Challenges in comprehensive molecular testing include complexities associated with the need to assess the presence of multiple variants, costs of running multiple sequential assays per sample, high assay quality control (QC) failure rates, clinical need for rapid turn-around time (TAT) to initiate therapy, and insufficient tissue samples. The ASPYRE-Lung NSCLC assay addresses gaps in multiplexed testing by simultaneously analyzing DNA and RNA, detecting 114 actionable genomic variants across 11 genes, consistent with current NSCLC treatment guidelines. This study was to assess the ease of adoption and performance of ASPYRE-Lung in third-party laboratories, comparing concordance across sites and with orthogonal methods.</p><p><strong>Methods: </strong>ASPYRE-Lung was established at two academic centers with multiple operators per site. Assay concordance was evaluated across three sites using 77 patient samples [61 derived from formalin-fixed paraffin-embedded (FFPE) tissue and 16 from cytology specimens].</p><p><strong>Results: </strong>Reproducibility for all 77 samples yielded a positive percent agreement (PPA) of 100% and negative percent agreement (NPA) of 99.99%. Concordance with next-generation sequencing (NGS)-based methods across all three sites was high with PPA of 97.2% and NPA of 99.96%.</p><p><strong>Conclusions: </strong>ASPYRE-Lung assay is a cost-effective, easy to adopt testing method requiring no specialized expertise or complicated bioinformatics, with the potential to inform genomic data on small tissue samples, thus enabling all patients with NSCLC to undergo biomarker testing in a timely manner and benefit from appropriate targeted therapies.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 11","pages":"3083-3095"},"PeriodicalIF":4.0,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11632423/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142819118","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}
Background: Pleural invasion (PI) is considered to be an adverse prognostic factor in non-small cell lung cancer (NSCLC). However, the prognostic roles of PI in pathologic (p)T3-4N0M0 NSCLC remain controversial. Therefore, this study aimed to evaluate the predictive value of PI in patients with pT3-4N0M0 NSCLC.
Methods: This study selected 9,185 patients with resected pT3-4N0M0 NSCLC from 2010 to 2019 from the Surveillance, Epidemiology, and End Results (SEER) database. PL0 was defined as lack of PI; PL1 and PL2 as invasion of the elastic layer and of the surface of the visceral pleural invasion (VPI), respectively; and PL3 as the parietal pleura invasion (PPI). Patients were divided into six groups according to PI status and T categories. This study used propensity score matching (PSM) to reduce baseline differences. The risk factors were statistically analyzed using the Cox proportional hazard model. Overall survival (OS) was assessed with the Kaplan-Meier method and log-rank test.
Results: Kaplan-Meier analysis stratified by T category showed worse OS in patients with PI (P<0.001). In multivariable Cox analysis of OS, patients with T3 and VPI had a significantly worse prognosis than did those with T3 but not PI (after PSM: P=0.001). There was no difference between those with T3 and VPI and those with T3 and PPI (after PSM: P=0.12) or between those with T3 PI and those with T4 but not PI (after PSM: P=0.30).
Conclusions: PI is a factor of poor prognosis in patients with pT3-4N0M0 NSCLC. Our results recommend future studies exploring the prognostic value of various T3-4 subcategories to allow PI to be separated from other T descriptors and confounders.
{"title":"Influence of pleural invasion on survival in pathologic T3-4N0M0 non-small cell lung cancer: a propensity score matching study based on the Surveillance, Epidemiology, and End Results database.","authors":"Wenqiang Zhang, Ziming Wang, Luyu Huang, Lara Sarigul, Diego Gonzalez-Rivas, Harun Badakhshi, Zhuangshi Huang, Jens-C Rueckert, Mahmoud Ismail","doi":"10.21037/tlcr-24-860","DOIUrl":"10.21037/tlcr-24-860","url":null,"abstract":"<p><strong>Background: </strong>Pleural invasion (PI) is considered to be an adverse prognostic factor in non-small cell lung cancer (NSCLC). However, the prognostic roles of PI in pathologic (p)T3-4N0M0 NSCLC remain controversial. Therefore, this study aimed to evaluate the predictive value of PI in patients with pT3-4N0M0 NSCLC.</p><p><strong>Methods: </strong>This study selected 9,185 patients with resected pT3-4N0M0 NSCLC from 2010 to 2019 from the Surveillance, Epidemiology, and End Results (SEER) database. PL0 was defined as lack of PI; PL1 and PL2 as invasion of the elastic layer and of the surface of the visceral pleural invasion (VPI), respectively; and PL3 as the parietal pleura invasion (PPI). Patients were divided into six groups according to PI status and T categories. This study used propensity score matching (PSM) to reduce baseline differences. The risk factors were statistically analyzed using the Cox proportional hazard model. Overall survival (OS) was assessed with the Kaplan-Meier method and log-rank test.</p><p><strong>Results: </strong>Kaplan-Meier analysis stratified by T category showed worse OS in patients with PI (P<0.001). In multivariable Cox analysis of OS, patients with T3 and VPI had a significantly worse prognosis than did those with T3 but not PI (after PSM: P=0.001). There was no difference between those with T3 and VPI and those with T3 and PPI (after PSM: P=0.12) or between those with T3 PI and those with T4 but not PI (after PSM: P=0.30).</p><p><strong>Conclusions: </strong>PI is a factor of poor prognosis in patients with pT3-4N0M0 NSCLC. Our results recommend future studies exploring the prognostic value of various T3-4 subcategories to allow PI to be separated from other T descriptors and confounders.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 11","pages":"3214-3223"},"PeriodicalIF":4.0,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11632439/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142819125","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}
Pub Date : 2024-11-30Epub Date: 2024-11-28DOI: 10.21037/tlcr-24-595
Chengyu Bian, Chenghao Fu, Wentao Xue, Yan Gu, Hongchang Wang, Wenhao Zhang, Guang Mu, Mei Yuan, Liang Chen, Jingjing Huang, Qianyun Wang, Jun Wang
Background: The precision of segmentectomy/subsegmentectomy for ground glass opacity (GGO)-dominant cT1a-bN0 non-small cell lung cancer (NSCLC), including mono-segmentectomy, mono-subsegmentectomy, combined subsegmentectomies, and single segmentectomy with adjacent subsegmentectomy, has improved. The aim of this study is to investigate their positional indications by focusing on the three-dimensional location of lesions, utilizing three-dimensional computed tomography bronchography and angiography (3D-CTBA).
Methods: We retrospectively analyzed 195 patients with GGO-dominant cT1a-bN0 NSCLC who underwent segmentectomy/subsegmentectomy between August 2015 and November 2020. We included 173 patients: mono-segmentectomy (71, 41.04%), mono-subsegmentectomy (37, 21.39%), combined subsegmentectomies (42, 24.28%), and single segmentectomy with adjacent subsegmentectomy (23,13.29%). Patient demographics and perioperative outcomes were compared among groups to identify positional indications.
Results: Significant differences were observed among the four groups in terms of lobe location of the lesions and their relationships with adjacent intersegmental veins (P<0.001), but not in their diameter and depth (P=0.33; P=0.79). All groups showed similar surgical margins (P=0.77) despite differences in the number of subsegments resected (P<0.001). No perioperative deaths or postoperative recurrences were reported. For lesions located in the middle region, located inter-segmentally, or with a diameter >1 cm, a greater number of subsegments were resected (P=0.02; P<0.001; P=0.003), while the surgical margins were not inferior to those located in the outer region, located intra-segmentally, or with a diameter ≤1 cm (P=0.29; P=0.77; P=0.46).
Conclusions: It is the specific lobe in which lesions are located and their relationship with adjacent intersegmental veins that determine the specific surgical procedure of segmentectomy/subsegmentectomy for GGO-dominant cT1a-bN0 NSCLC, rather than their diameter and depth.
{"title":"Three-dimensional reconstruction for determining positional indications of pulmonary segmentectomy/subsegmentectomy for ground glass opacity-dominant clinical T1a-bN0 non-small cell lung cancer.","authors":"Chengyu Bian, Chenghao Fu, Wentao Xue, Yan Gu, Hongchang Wang, Wenhao Zhang, Guang Mu, Mei Yuan, Liang Chen, Jingjing Huang, Qianyun Wang, Jun Wang","doi":"10.21037/tlcr-24-595","DOIUrl":"10.21037/tlcr-24-595","url":null,"abstract":"<p><strong>Background: </strong>The precision of segmentectomy/subsegmentectomy for ground glass opacity (GGO)-dominant cT1a-bN0 non-small cell lung cancer (NSCLC), including mono-segmentectomy, mono-subsegmentectomy, combined subsegmentectomies, and single segmentectomy with adjacent subsegmentectomy, has improved. The aim of this study is to investigate their positional indications by focusing on the three-dimensional location of lesions, utilizing three-dimensional computed tomography bronchography and angiography (3D-CTBA).</p><p><strong>Methods: </strong>We retrospectively analyzed 195 patients with GGO-dominant cT1a-bN0 NSCLC who underwent segmentectomy/subsegmentectomy between August 2015 and November 2020. We included 173 patients: mono-segmentectomy (71, 41.04%), mono-subsegmentectomy (37, 21.39%), combined subsegmentectomies (42, 24.28%), and single segmentectomy with adjacent subsegmentectomy (23,13.29%). Patient demographics and perioperative outcomes were compared among groups to identify positional indications.</p><p><strong>Results: </strong>Significant differences were observed among the four groups in terms of lobe location of the lesions and their relationships with adjacent intersegmental veins (P<0.001), but not in their diameter and depth (P=0.33; P=0.79). All groups showed similar surgical margins (P=0.77) despite differences in the number of subsegments resected (P<0.001). No perioperative deaths or postoperative recurrences were reported. For lesions located in the middle region, located inter-segmentally, or with a diameter >1 cm, a greater number of subsegments were resected (P=0.02; P<0.001; P=0.003), while the surgical margins were not inferior to those located in the outer region, located intra-segmentally, or with a diameter ≤1 cm (P=0.29; P=0.77; P=0.46).</p><p><strong>Conclusions: </strong>It is the specific lobe in which lesions are located and their relationship with adjacent intersegmental veins that determine the specific surgical procedure of segmentectomy/subsegmentectomy for GGO-dominant cT1a-bN0 NSCLC, rather than their diameter and depth.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 11","pages":"2947-2957"},"PeriodicalIF":4.0,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11632417/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142819205","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}
Background: The third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) osimertinib is widely used as a first-line treatment for EGFR-mutated non-small cell lung cancer (NSCLC). However, there is no established treatment for osimertinib resistance, so second-generation afatinib is an alternative treatment option. The purpose of this study was to elucidate gene alterations associated with afatinib efficacy and resistance by analyzing cell-free DNA (cfDNA) obtained from patients with EGFR-mutated NSCLC.
Methods: This study was conducted as a prospective clinical trial in patients with EGFR-mutated NSCLC at multiple institutions. We analyzed plasma cfDNA from the patients treated with afatinib as the first-line therapy.
Results: Paired specimens were obtained before and at the time of resistance to afatinib treatment, and specimens only at afatinib resistance were obtained from 22 and 18 patients, respectively. In plasma cfDNA from the 22 cases, driver EGFR mutations were detected in 13 cases (59.1%), and the compound V834L mutation was detected in two cases in cis with EGFR-L858R mutation. The median progression-free survival (mPFS) was remarkably shorter in patients with V834L than in all 22 cases (4.2 vs. 9.2 months). Moreover, we detected V834L and T790M combined with EGFR-L858R in the cfDNA from one patient resistant to afatinib. Preclinical experiments using EGFR-L858R, with or without V834L, in Ba/F3 cells revealed that V834L with L858R conferred resistance to low concentrations of EGFR-TKIs, including afatinib and osimertinib. In three cases of EGFR-L858R+V834L, other co-mutations, including TP53, CTNNB1, and RB1, were detected either before or after afatinib resistance.
Conclusions: These results suggested that V834L cooperates with other coexisting mutations to influence the therapeutic efficacy of EGFR-TKIs.
{"title":"<i>EGFR-V834L</i> combined with L858R mutation reduced afatinib sensitivity and associated to early recurrence in lung cancer.","authors":"Nanao Terada, Hayato Koba, Shigeki Nanjo, Hideharu Kimura, Akihiro Nishiyama, Taro Yoneda, Takayuki Takeda, Hiroki Shirasaki, Koichi Nishi, Takashi Sone, Tadaaki Yamada, Koichi Takayama, Kazuhiko Shibata, Hiroki Matsuoka, Toshiyuki Kita, Kazuhiro Nagata, Yuichi Tambo, Noriyuki Ohkura, Johsuke Hara, Kazuo Kasahara, Shinji Kohsaka, Hiroyuki Mano, Seiji Yano","doi":"10.21037/tlcr-24-471","DOIUrl":"10.21037/tlcr-24-471","url":null,"abstract":"<p><strong>Background: </strong>The third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) osimertinib is widely used as a first-line treatment for <i>EGFR</i>-mutated non-small cell lung cancer (NSCLC). However, there is no established treatment for osimertinib resistance, so second-generation afatinib is an alternative treatment option. The purpose of this study was to elucidate gene alterations associated with afatinib efficacy and resistance by analyzing cell-free DNA (cfDNA) obtained from patients with <i>EGFR</i>-mutated NSCLC.</p><p><strong>Methods: </strong>This study was conducted as a prospective clinical trial in patients with <i>EGFR</i>-mutated NSCLC at multiple institutions. We analyzed plasma cfDNA from the patients treated with afatinib as the first-line therapy.</p><p><strong>Results: </strong>Paired specimens were obtained before and at the time of resistance to afatinib treatment, and specimens only at afatinib resistance were obtained from 22 and 18 patients, respectively. In plasma cfDNA from the 22 cases, driver <i>EGFR</i> mutations were detected in 13 cases (59.1%), and the compound V834L mutation was detected in two cases in cis with <i>EGFR</i>-L858R mutation. The median progression-free survival (mPFS) was remarkably shorter in patients with V834L than in all 22 cases (4.2 <i>vs</i>. 9.2 months). Moreover, we detected V834L and T790M combined with <i>EGFR</i>-L858R in the cfDNA from one patient resistant to afatinib. Preclinical experiments using <i>EGFR</i>-L858R, with or without V834L, in Ba/F3 cells revealed that V834L with L858R conferred resistance to low concentrations of EGFR-TKIs, including afatinib and osimertinib. In three cases of <i>EGFR</i>-L858R+V834L, other co-mutations, including <i>TP53</i>, <i>CTNNB1</i>, and <i>RB1</i>, were detected either before or after afatinib resistance.</p><p><strong>Conclusions: </strong>These results suggested that V834L cooperates with other coexisting mutations to influence the therapeutic efficacy of EGFR-TKIs.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 11","pages":"3067-3082"},"PeriodicalIF":4.0,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11632430/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142818384","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}
Pub Date : 2024-11-30Epub Date: 2024-11-14DOI: 10.21037/tlcr-24-433
Angelika Terbuch, Selma Konjic, Verena Schlintl, Gudrun Absenger, Philipp J Jost, Jörg Lindenmann, Paul Swatek, Nikolaus John, Teresa John, Robert Wurm, Martin Zacharias, Florian Posch, Maximilian J Hochmair, Hannah Fabikan, Christoph Weinlinger, Oliver Illini, Lena Horvath, Gabriele Gamerith, Dominik Wolf, Florian Augustin, Luka Brcic
Background: Apart from ALK fusions and the common EGFR mutations, targetable molecular alterations are irrelevant for adjuvant treatment decision making in early-stage non-small cell lung cancer (NSCLC). This retrospective analysis aimed to investigate if there is a difference in recurrence-free survival in stage I-III NSCLC harboring druggable molecular alterations compared to subtypes without targetable molecular alterations.
Methods: All consecutive patients who underwent surgery with curative intent for NSCLC (stage I-III) with targetable mutations between January 2015 and December 2020 at three Austrian institutions were identified and compared with tumors without targetable molecular alterations. Tumors with the EGFR-mutated subtype were excluded due to already existing results from prospective trials.
Results: One hundred and sixty subjects had tumors with molecular alterations and 355 subjects served as control cohort. There was a higher prevalence of female sex (P<0.001) and never-smokers (P=0.01) among patients with tumors harboring oncogenic driver mutations. The three most common alterations were the KRAS G12C mutation (n=92), ALK fusions (n=21), and the BRAF V600E mutation (n=15). The 1-, 3- and 5-year cumulative incidence of recurrence estimates were 16%, 38% and 46% in patients without molecular alterations and 16%, 38% and 48% in patients with the KRAS G12C mutation and 12%, 33% and 55% in patients with other molecular alterations, respectively (P=0.89). Univariable predictors of an increased recurrence risk were higher tumor stage (P<0.001), receipt of neoadjuvant treatment (P<0.001) and receipt of adjuvant treatment (P=0.03). The lack of association between molecular alteration status and recurrence risk prevailed after multivariable adjustment for tumor stage and perioperative treatment (P=0.82 for KRAS G12C mutation and P=0.43 for any other molecular alteration).
Conclusions: NSCLC patients with resected tumors that harbor molecular alterations have the same recurrence risk as patients with tumors without molecular alterations if treated with surgery plus chemotherapy when indicated.
{"title":"Prognostic impact of targetable driver alterations in resected early-stage lung cancer.","authors":"Angelika Terbuch, Selma Konjic, Verena Schlintl, Gudrun Absenger, Philipp J Jost, Jörg Lindenmann, Paul Swatek, Nikolaus John, Teresa John, Robert Wurm, Martin Zacharias, Florian Posch, Maximilian J Hochmair, Hannah Fabikan, Christoph Weinlinger, Oliver Illini, Lena Horvath, Gabriele Gamerith, Dominik Wolf, Florian Augustin, Luka Brcic","doi":"10.21037/tlcr-24-433","DOIUrl":"10.21037/tlcr-24-433","url":null,"abstract":"<p><strong>Background: </strong>Apart from <i>ALK</i> fusions and the common <i>EGFR</i> mutations, targetable molecular alterations are irrelevant for adjuvant treatment decision making in early-stage non-small cell lung cancer (NSCLC). This retrospective analysis aimed to investigate if there is a difference in recurrence-free survival in stage I-III NSCLC harboring druggable molecular alterations compared to subtypes without targetable molecular alterations.</p><p><strong>Methods: </strong>All consecutive patients who underwent surgery with curative intent for NSCLC (stage I-III) with targetable mutations between January 2015 and December 2020 at three Austrian institutions were identified and compared with tumors without targetable molecular alterations. Tumors with the <i>EGFR</i>-mutated subtype were excluded due to already existing results from prospective trials.</p><p><strong>Results: </strong>One hundred and sixty subjects had tumors with molecular alterations and 355 subjects served as control cohort. There was a higher prevalence of female sex (P<0.001) and never-smokers (P=0.01) among patients with tumors harboring oncogenic driver mutations. The three most common alterations were the <i>KRAS G12C</i> mutation (n=92), <i>ALK</i> fusions (n=21), and the <i>BRAF V600E</i> mutation (n=15). The 1-, 3- and 5-year cumulative incidence of recurrence estimates were 16%, 38% and 46% in patients without molecular alterations and 16%, 38% and 48% in patients with the <i>KRAS G12C</i> mutation and 12%, 33% and 55% in patients with other molecular alterations, respectively (P=0.89). Univariable predictors of an increased recurrence risk were higher tumor stage (P<0.001), receipt of neoadjuvant treatment (P<0.001) and receipt of adjuvant treatment (P=0.03). The lack of association between molecular alteration status and recurrence risk prevailed after multivariable adjustment for tumor stage and perioperative treatment (P=0.82 for <i>KRAS G12C</i> mutation and P=0.43 for any other molecular alteration).</p><p><strong>Conclusions: </strong>NSCLC patients with resected tumors that harbor molecular alterations have the same recurrence risk as patients with tumors without molecular alterations if treated with surgery plus chemotherapy when indicated.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 11","pages":"3096-3105"},"PeriodicalIF":4.0,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11632426/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142819169","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}
Pub Date : 2024-11-30Epub Date: 2024-11-28DOI: 10.21037/tlcr-24-570
Gerhard Hamilton, Sandra Stickler, Mikhail Ermakov, Marie-Therese Eggerstorfer, Francesca Paola Nocera, Martin Hohenegger, Lukas Weigl, Maximilian Johannes Hochmair, Karl Kashofer
Background: Approximately 30% of the non-small cell lung cancer (NSCLC) patients which harbor no recognizable oncogenic driver mutation are not eligible for targeted therapy. Functional drug screening of tumor cells helps to identify susceptible drug targets not recognized by gene panels for targeted mutation analysis. The aim of this study is to characterize the BH1406 cell line carrying an activating SOS1 mutation and to check its sensitivity to cognate inhibitors.
Methods: The NSCLC cell line BH1406 was established from a pleural effusion and found to be sensitive to the SOS1 inhibitor BAY-293 in initial viability screenings. Since in a limited next-generation sequencing (NGS) lung cancer mutation panel no driver could be detected, the patient underwent chemotherapy with poor outcome. This cell line was further characterized by exome sequencing, SOS1 Western blotting, comparison of two-dimensional (2D) and three-dimensional (3D) chemosensitivity assays and phosphoprotein arrays.
Results: In whole-exome sequencing (WES) the SOS1 mutation P481delinsLFFL, positioned near the known P478L activating mutation was detected. Besides BAY-293, BH1406 cells proved to be sensitive to the SOS1 inhibitors MRTX0902 and BI-3406. The sensitivity of BH1406 cells to BI-3406 was increased under 3D conditions compared to 2D cultures. Western blot phosphoprotein arrays revealed reduced phosphorylation of CREB, GSK3, CHK-2 and STAT3 in BH1406 by BAY-293 treatment in 2D culture. In 3D conditions, cells switched from GSK3α to elevated ERK1/2 signaling, again blocked by the SOS1 inhibitor BAY-293. Similar results were obtained for the SOS1 inhibitors MRTX0902 and BI3406. Additionally, the PI3K inhibitor dactolisib, the GSK-3 inhibitor BI-5521 as well as the bromodomain protein-directed PROTAC ARV-771 inhibited the growth of BH1406 cells significantly and showed synergistic interaction with BAY-293. Furthermore, Western blots demonstrated reduced expression of SOS1 and MYC proteins in response to BAY-293 treatment.
Conclusions: The rare SOS1 P481delinsLFFL mutation in lung cancer may be targetable with corresponding inhibitors, alone or in combination with GSK3/PI3K/BET inhibitors. BH1406 cells represent a novel cellular model suitable for the molecular characterization of SOS1 druggability. Such rare oncogenic driver genes are not included in standard NGS panels and need to be detected by expanded assays like WES.
背景:大约30%没有可识别的致癌驱动突变的非小细胞肺癌(NSCLC)患者不适合靶向治疗。肿瘤细胞的功能药物筛选有助于识别基因面板无法识别的敏感药物靶点,用于靶向突变分析。本研究的目的是表征携带激活SOS1突变的BH1406细胞系,并检查其对同源抑制剂的敏感性。方法:从胸腔积液中建立非小细胞肺癌细胞系BH1406,在初始活力筛选中发现对SOS1抑制剂BAY-293敏感。由于在有限的下一代测序(NGS)肺癌突变面板中没有检测到驱动因素,因此患者接受了化疗,结果不佳。该细胞系进一步通过外显子组测序、SOS1 Western blotting、二维(2D)和三维(3D)化学敏感性比较和磷酸化蛋白阵列进行表征。结果:在全外显子组测序(WES)中,检测到位于已知P478L激活突变附近的SOS1突变P481delinsLFFL。除BAY-293外,BH1406细胞还对SOS1抑制剂MRTX0902和BI-3406敏感。与2D培养相比,BH1406细胞在3D条件下对BI-3406的敏感性增加。Western blot磷酸化蛋白阵列显示BAY-293在2D培养中降低BH1406中CREB、GSK3、CHK-2和STAT3的磷酸化水平。在3D条件下,细胞从GSK3α切换到升高的ERK1/2信号,再次被SOS1抑制剂BAY-293阻断。SOS1抑制剂MRTX0902和BI3406也获得了类似的结果。此外,PI3K抑制剂dactolisib、GSK-3抑制剂BI-5521以及bromodomain蛋白导向的PROTAC ARV-771均能显著抑制BH1406细胞的生长,并与BAY-293表现出协同作用。此外,Western blot结果显示,BAY-293治疗后SOS1和MYC蛋白的表达降低。结论:肺癌中罕见的SOS1 P481delinsLFFL突变可能与相应的抑制剂靶向,单独或联合GSK3/PI3K/BET抑制剂。BH1406细胞是一种适合SOS1药物性分子表征的新型细胞模型。这种罕见的致癌驱动基因不包括在标准的NGS面板中,需要通过像WES这样的扩展试验来检测。
{"title":"Characterization of the BH1406 non-small cell lung cancer (NSCLC) cell line carrying an activating SOS1 mutation.","authors":"Gerhard Hamilton, Sandra Stickler, Mikhail Ermakov, Marie-Therese Eggerstorfer, Francesca Paola Nocera, Martin Hohenegger, Lukas Weigl, Maximilian Johannes Hochmair, Karl Kashofer","doi":"10.21037/tlcr-24-570","DOIUrl":"10.21037/tlcr-24-570","url":null,"abstract":"<p><strong>Background: </strong>Approximately 30% of the non-small cell lung cancer (NSCLC) patients which harbor no recognizable oncogenic driver mutation are not eligible for targeted therapy. Functional drug screening of tumor cells helps to identify susceptible drug targets not recognized by gene panels for targeted mutation analysis. The aim of this study is to characterize the BH1406 cell line carrying an activating SOS1 mutation and to check its sensitivity to cognate inhibitors.</p><p><strong>Methods: </strong>The NSCLC cell line BH1406 was established from a pleural effusion and found to be sensitive to the SOS1 inhibitor BAY-293 in initial viability screenings. Since in a limited next-generation sequencing (NGS) lung cancer mutation panel no driver could be detected, the patient underwent chemotherapy with poor outcome. This cell line was further characterized by exome sequencing, SOS1 Western blotting, comparison of two-dimensional (2D) and three-dimensional (3D) chemosensitivity assays and phosphoprotein arrays.</p><p><strong>Results: </strong>In whole-exome sequencing (WES) the SOS1 mutation P481delinsLFFL, positioned near the known P478L activating mutation was detected. Besides BAY-293, BH1406 cells proved to be sensitive to the SOS1 inhibitors MRTX0902 and BI-3406. The sensitivity of BH1406 cells to BI-3406 was increased under 3D conditions compared to 2D cultures. Western blot phosphoprotein arrays revealed reduced phosphorylation of CREB, GSK3, CHK-2 and STAT3 in BH1406 by BAY-293 treatment in 2D culture. In 3D conditions, cells switched from GSK3α to elevated ERK1/2 signaling, again blocked by the SOS1 inhibitor BAY-293. Similar results were obtained for the SOS1 inhibitors MRTX0902 and BI3406. Additionally, the PI3K inhibitor dactolisib, the GSK-3 inhibitor BI-5521 as well as the bromodomain protein-directed PROTAC ARV-771 inhibited the growth of BH1406 cells significantly and showed synergistic interaction with BAY-293. Furthermore, Western blots demonstrated reduced expression of SOS1 and MYC proteins in response to BAY-293 treatment.</p><p><strong>Conclusions: </strong>The rare SOS1 P481delinsLFFL mutation in lung cancer may be targetable with corresponding inhibitors, alone or in combination with GSK3/PI3K/BET inhibitors. BH1406 cells represent a novel cellular model suitable for the molecular characterization of SOS1 druggability. Such rare oncogenic driver genes are not included in standard NGS panels and need to be detected by expanded assays like WES.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 11","pages":"2987-2997"},"PeriodicalIF":4.0,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11632420/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142818823","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}
Background: Delineating gross tumor volume (GTV) using computed tomography (CT) imaging is the standard for lung cancer contouring, but discrepancies among observers compromise accuracy and reliability. Magnetic resonance imaging (MRI) provides superior soft-tissue resolution compared to CT, thus, we design this retrospective study to compare the treatment outcomes of magnetic resonance-based (MR-based) and CT-based tumor delineation in locally advanced non-small cell lung cancer (LA-NSCLC) patients treated with hypo-fractionated concurrent chemoradiotherapy (hypo-CCRT).
Methods: A total of 293 LA-NSCLC patients treated with hypo-CCRT from three trials between October 2015 and October 2020 were screened. Ninety patients with each MR-based delineation and CT-based delineation of the primary tumor were selected for analysis. In the MR-based delineation group, T1-enhanced MR images was rigidly registered with 10 respiratory phases of planning CT images, respectively. The primary tumors were contoured on each respiratory phase based on co-registered MRI. The locoregional progression-free survival (LPFS), progression-free survival (PFS), overall survival (OS) and toxicities in both groups were analyzed.
Results: The 2-year LPFS rate was 69.2% [95% confidence interval (CI): 59.6-80.2%] in the MR-based delineation group and 61.0% (95% CI: 50.9-73.0%) in the CT-based delineation group (P=0.37). There was no significant difference in median PFS (P=0.45) or OS (P=0.69) between the two groups. The MR-based delineation group had smaller planning target volume (186.1 vs. 315.3 cm3, P<0.001), lower incidences of ≥G2 pneumonitis (10% vs. 24.4%, P=0.001) and ≥G3 esophagitis (2.2% vs. 15.6%, P<0.001). In evaluating the patterns of recurrence, in-field recurrences were the dominant type in both groups (21 out of 27 patients in MR-based delineation group, 24 out of 32 patients in CT-based delineation group).
Conclusions: MR-based delineation in hypo-CCRT was feasible and achieved similar treatment efficacy to CT-based delineation. The use of MR imaging to reduce the target volume resulted in promising local control and lower incidence of radiation-induced toxicities.
背景:使用计算机断层扫描(CT)成像描绘肿瘤总体积(GTV)是肺癌轮廓的标准,但观察者之间的差异损害了准确性和可靠性。与CT相比,磁共振成像(MRI)提供了更好的软组织分辨率,因此,我们设计了这项回顾性研究,以比较基于磁共振(mr)和基于CT的局部晚期非小细胞肺癌(LA-NSCLC)患者接受低分级并行放化疗(hypo-CCRT)治疗的治疗结果。方法:对2015年10月至2020年10月期间接受低ccrt治疗的293例LA-NSCLC患者进行筛查。分别对原发肿瘤进行mri和ct描述的90例患者进行分析。在基于MR的圈定组中,t1增强MR图像分别与规划CT图像的10个呼吸期严格配准。在共登记MRI的基础上,在每个呼吸期绘制原发肿瘤的轮廓。分析两组患者的局部无进展生存期(LPFS)、无进展生存期(PFS)、总生存期(OS)及毒副反应。结果:基于mri的划定组2年LPFS率为69.2%[95%可信区间(CI): 59.6-80.2%],基于ct的划定组为61.0% (95% CI: 50.9-73.0%) (P=0.37)。两组间的中位PFS (P=0.45)和OS (P=0.69)无显著差异。基于mr的划定组计划靶体积较小(186.1 vs. 315.3 cm3, vs. 24.4%, P=0.001),≥G3级食管炎(2.2% vs. 15.6%, P)。结论:基于mr的划定在低ccrt中是可行的,其治疗效果与基于ct的划定相似。使用磁共振成像来缩小靶体积导致有希望的局部控制和降低辐射引起的毒性的发生率。
{"title":"Comparing the outcomes of MR-based versus CT-based tumor delineation in locally advanced non-small cell lung cancer treated with hypo-fractionated radiotherapy and concurrent chemotherapy.","authors":"Pengxin Zhang, Shouliang Ding, Kangqiang Peng, Haoqiang He, Daquan Wang, Rui Zhou, Bin Wang, Jinyu Guo, Hongdong Liu, Xiaoyan Huang, Chuanmiao Xie, Hui Liu, Bo Qiu","doi":"10.21037/tlcr-24-341","DOIUrl":"10.21037/tlcr-24-341","url":null,"abstract":"<p><strong>Background: </strong>Delineating gross tumor volume (GTV) using computed tomography (CT) imaging is the standard for lung cancer contouring, but discrepancies among observers compromise accuracy and reliability. Magnetic resonance imaging (MRI) provides superior soft-tissue resolution compared to CT, thus, we design this retrospective study to compare the treatment outcomes of magnetic resonance-based (MR-based) and CT-based tumor delineation in locally advanced non-small cell lung cancer (LA-NSCLC) patients treated with hypo-fractionated concurrent chemoradiotherapy (hypo-CCRT).</p><p><strong>Methods: </strong>A total of 293 LA-NSCLC patients treated with hypo-CCRT from three trials between October 2015 and October 2020 were screened. Ninety patients with each MR-based delineation and CT-based delineation of the primary tumor were selected for analysis. In the MR-based delineation group, T1-enhanced MR images was rigidly registered with 10 respiratory phases of planning CT images, respectively. The primary tumors were contoured on each respiratory phase based on co-registered MRI. The locoregional progression-free survival (LPFS), progression-free survival (PFS), overall survival (OS) and toxicities in both groups were analyzed.</p><p><strong>Results: </strong>The 2-year LPFS rate was 69.2% [95% confidence interval (CI): 59.6-80.2%] in the MR-based delineation group and 61.0% (95% CI: 50.9-73.0%) in the CT-based delineation group (P=0.37). There was no significant difference in median PFS (P=0.45) or OS (P=0.69) between the two groups. The MR-based delineation group had smaller planning target volume (186.1 <i>vs.</i> 315.3 cm<sup>3</sup>, P<0.001), lower incidences of ≥G2 pneumonitis (10% <i>vs.</i> 24.4%, P=0.001) and ≥G3 esophagitis (2.2% <i>vs.</i> 15.6%, P<0.001). In evaluating the patterns of recurrence, in-field recurrences were the dominant type in both groups (21 out of 27 patients in MR-based delineation group, 24 out of 32 patients in CT-based delineation group).</p><p><strong>Conclusions: </strong>MR-based delineation in hypo-CCRT was feasible and achieved similar treatment efficacy to CT-based delineation. The use of MR imaging to reduce the target volume resulted in promising local control and lower incidence of radiation-induced toxicities.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 11","pages":"2890-2902"},"PeriodicalIF":4.0,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11632429/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142819003","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}
Background: Non-small cell lung cancer (NSCLC), the most prevalent lung cancer subtype, presents significant treatment challenges. Cisplatin (CP)-based regimens are central to the treatment of multiple solid tumors, but its use is restricted due to its dose-related renal toxicity. We previously found that fluorofenidone {1-[3-fluorophenyl]-5-methyl-2-[(1H)]-pyridone (AKF-PD)} effectively reverses CP-induced acute kidney injury (AKI). However, it remains unclear whether AKF-PD can synergistically ameliorate NSCLC when used together with CP. Thus, this study sought to investigate the effect of AKF-PD on NSCLC and examined its combinatory use with CP for cancer treatment.
Methods: We conducted cell viability assays, 5-ethynyl-2'-deoxyuridine (EdU) experiments, colony-forming assays, wound-healing tests, and Transwell experiments in A549 and H1299 cells to explore the effects of AKF-PD on NSCLC. We then detected the epithelial-mesenchymal transition (EMT) markers [i.e., epithelial cadherin (E-cadherin), matrix metallopeptidase 9 (MMP9), vimentin, and snail family transcriptional repressor 1 (SNAIL)], phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mechanistic target of rapamycin (mTOR), and mitogen-activated protein kinase (MAPK), to identify the potential mechanisms of AKF-PD. Further, via the combined use of AKF-PD and CP, we found that AKF-PD enhanced the antitumor effect of CP, and we suggest that this may be due to its inhibitory effect on EMT. We also examined the effect of combining AKF-PD and CP in other cancer cell lines, including Hela, SiHA, MDA-MB-231, 5-8F, and UM-UC-3 cells.
Results: AKF-PD significantly inhibited the proliferation and invasion of NSCLC cells (A549 and H1299), suppressed the activation of the MAPK and PI3K/AKT/mTOR pathways, and inhibited the EMT of the tumor cells. When AKF-PD was used in combination with CP, these effects were further enhanced. We also found that AKF-PD enhanced the anti-cancer effect of CP in a variety of cancer cell lines, including cervical cancer (Hela cells and SiHA cells), nasopharyngeal cancer (5-8F cells), triple-negative breast cancer (MDA-MB-231 cells), and bladder cancer (UM-UC-3 cells).
Conclusions: AKF-PD not only mitigates CP-induced AKI but also enhances the anti-cancer efficacy of CP. Our findings provide valuable insights into the treatment of NSCLC and may have clinical applications.
{"title":"Fluorofenidone enhances cisplatin efficacy in non-small cell lung cancer: a novel approach to inhibiting cancer progression.","authors":"Shunjun Wang, Guowei Liu, Laishun Yu, Chenzi Zhang, Fabrizio Marcucci, Yupeng Jiang","doi":"10.21037/tlcr-24-811","DOIUrl":"10.21037/tlcr-24-811","url":null,"abstract":"<p><strong>Background: </strong>Non-small cell lung cancer (NSCLC), the most prevalent lung cancer subtype, presents significant treatment challenges. Cisplatin (CP)-based regimens are central to the treatment of multiple solid tumors, but its use is restricted due to its dose-related renal toxicity. We previously found that fluorofenidone {1-[3-fluorophenyl]-5-methyl-2-[(1H)]-pyridone (AKF-PD)} effectively reverses CP-induced acute kidney injury (AKI). However, it remains unclear whether AKF-PD can synergistically ameliorate NSCLC when used together with CP. Thus, this study sought to investigate the effect of AKF-PD on NSCLC and examined its combinatory use with CP for cancer treatment.</p><p><strong>Methods: </strong>We conducted cell viability assays, 5-ethynyl-2'-deoxyuridine (EdU) experiments, colony-forming assays, wound-healing tests, and Transwell experiments in A549 and H1299 cells to explore the effects of AKF-PD on NSCLC. We then detected the epithelial-mesenchymal transition (EMT) markers [i.e., epithelial cadherin (E-cadherin), matrix metallopeptidase 9 (MMP9), vimentin, and snail family transcriptional repressor 1 (SNAIL)], phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mechanistic target of rapamycin (mTOR), and mitogen-activated protein kinase (MAPK), to identify the potential mechanisms of AKF-PD. Further, via the combined use of AKF-PD and CP, we found that AKF-PD enhanced the antitumor effect of CP, and we suggest that this may be due to its inhibitory effect on EMT. We also examined the effect of combining AKF-PD and CP in other cancer cell lines, including Hela, SiHA, MDA-MB-231, 5-8F, and UM-UC-3 cells.</p><p><strong>Results: </strong>AKF-PD significantly inhibited the proliferation and invasion of NSCLC cells (A549 and H1299), suppressed the activation of the MAPK and PI3K/AKT/mTOR pathways, and inhibited the EMT of the tumor cells. When AKF-PD was used in combination with CP, these effects were further enhanced. We also found that AKF-PD enhanced the anti-cancer effect of CP in a variety of cancer cell lines, including cervical cancer (Hela cells and SiHA cells), nasopharyngeal cancer (5-8F cells), triple-negative breast cancer (MDA-MB-231 cells), and bladder cancer (UM-UC-3 cells).</p><p><strong>Conclusions: </strong>AKF-PD not only mitigates CP-induced AKI but also enhances the anti-cancer efficacy of CP. Our findings provide valuable insights into the treatment of NSCLC and may have clinical applications.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 11","pages":"3175-3188"},"PeriodicalIF":4.0,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11632444/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142819122","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}
Pub Date : 2024-11-30Epub Date: 2024-11-18DOI: 10.21037/tlcr-24-537
Beong Ki Kim, Ye Seul Seong, Se Hyun Kwak, Eun Hye Lee, Sang Hoon Lee, Eun Young Kim, Yoon Soo Chang, Chi Young Kim
Background: The introduction of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) has revolutionized advanced non-small cell lung cancer (NSCLC) treatment. However, their efficacy can be compromised by concurrent use of gastric acid suppressants (GASs), such as proton pump inhibitors (PPIs) and histamine 2 receptor antagonists (H2RAs). This study aimed to update the evidence on the impact of GASs on the overall survival (OS) and progression-free survival (PFS) in patients on EGFR-TKI treatment.
Methods: A systematic review and meta-analysis were conducted using data from PubMed, Embase, Cochrane Library, Web of Science, Scopus, KoreaMed, and preprint repositories. Data from 13 retrospective studies, involving 10,814 patients, were analyzed.
Results: Overall, 34.6% of the patients used GASs, with most being Asian females and non-smokers. Most patients had EGFR-mutated adenocarcinoma, reflecting typical EGFR-TKI usage scenarios. Concurrent use of GASs was significantly associated with reduced OS [hazard ratio (HR) =1.34, 95% confidence interval (CI): 1.26-1.42], and PFS (HR =1.52, 95% CI: 1.25-1.86). In subgroup analysis, PPIs had a more negative impact on OS (HR =1.64, 95% CI: 1.51-1.79) than did H2RAs (HR =1.11, 95% CI: 0.95-1.31). Longer overlap times of GASs correlated with a higher trend in HRs for OS. However, the results for PFS were not significant in both subgroup analyses.
Conclusions: Concurrent use of GASs with EGFR-TKIs is linked to poorer OS and PFS in patients with advanced NSCLC. Careful consideration is advised when prescribing GASs, including adjusting administration timing, minimizing overlap duration, or opting for H2RAs over PPIs. Further research is needed to optimize treatment protocols, specifically addressing the duration of overlap time, to improve patient outcomes.
{"title":"Optimizing epidermal growth factor receptor-tyrosine kinase inhibitor treatment in lung cancer: a systematic review and meta-analysis of the influence of gastric acid suppressants.","authors":"Beong Ki Kim, Ye Seul Seong, Se Hyun Kwak, Eun Hye Lee, Sang Hoon Lee, Eun Young Kim, Yoon Soo Chang, Chi Young Kim","doi":"10.21037/tlcr-24-537","DOIUrl":"10.21037/tlcr-24-537","url":null,"abstract":"<p><strong>Background: </strong>The introduction of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) has revolutionized advanced non-small cell lung cancer (NSCLC) treatment. However, their efficacy can be compromised by concurrent use of gastric acid suppressants (GASs), such as proton pump inhibitors (PPIs) and histamine 2 receptor antagonists (H2RAs). This study aimed to update the evidence on the impact of GASs on the overall survival (OS) and progression-free survival (PFS) in patients on EGFR-TKI treatment.</p><p><strong>Methods: </strong>A systematic review and meta-analysis were conducted using data from PubMed, Embase, Cochrane Library, Web of Science, Scopus, KoreaMed, and preprint repositories. Data from 13 retrospective studies, involving 10,814 patients, were analyzed.</p><p><strong>Results: </strong>Overall, 34.6% of the patients used GASs, with most being Asian females and non-smokers. Most patients had EGFR-mutated adenocarcinoma, reflecting typical EGFR-TKI usage scenarios. Concurrent use of GASs was significantly associated with reduced OS [hazard ratio (HR) =1.34, 95% confidence interval (CI): 1.26-1.42], and PFS (HR =1.52, 95% CI: 1.25-1.86). In subgroup analysis, PPIs had a more negative impact on OS (HR =1.64, 95% CI: 1.51-1.79) than did H2RAs (HR =1.11, 95% CI: 0.95-1.31). Longer overlap times of GASs correlated with a higher trend in HRs for OS. However, the results for PFS were not significant in both subgroup analyses.</p><p><strong>Conclusions: </strong>Concurrent use of GASs with EGFR-TKIs is linked to poorer OS and PFS in patients with advanced NSCLC. Careful consideration is advised when prescribing GASs, including adjusting administration timing, minimizing overlap duration, or opting for H2RAs over PPIs. Further research is needed to optimize treatment protocols, specifically addressing the duration of overlap time, to improve patient outcomes.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 11","pages":"2934-2946"},"PeriodicalIF":4.0,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11632441/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142819156","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}