1
|
Zhao Y, An D, Bi L. Effect of Co-Administration of Midazolam and Dexmedetomidine on Haemodynamics and Stress Response in Elderly Patients with Non-Small Cell Lung Cancer. J INVEST SURG 2025; 38:2445587. [PMID: 39756799 DOI: 10.1080/08941939.2024.2445587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 10/11/2024] [Accepted: 12/16/2024] [Indexed: 01/07/2025]
Abstract
OBJECTIVE This study aimed to evaluate the effect of co-administration of midazolam and dexmedetomidine on hemodynamics and stress response in elderly patients with non-small cell lung cancer (NSCLC). METHODS In this prospective, randomized controlled trial, 154 elderly NSCLC patients scheduled for lobectomy in our oncology department from January 2019 to December 2021 were recruited. Patients were randomized 1:1 to receive either dexmedetomidine (control group) or dexmedetomidine plus midazolam (study group) for anesthesia during lobectomy via the random number table method, with 77 patients in each group. Perioperative indicators, hemodynamics, and stress reactions of the patients were recorded and compared between the two groups to investigate the efficacy of the two different anesthetic protocols. RESULTS No significant differences were observed between the two groups in terms of operative time, anesthesia time, and intraoperative bleeding volume (p > 0.05). Preoperative pain, pain at anesthesia recovery, and pain levels 7 days postoperatively were also comparable between the two groups. In the study group, the awakening time was 15 ± 2 min significantly shorter compared to the control group (25 ± 3 min). Cooperation within the first hour was significantly faster by 8.5 ± 0.5 min compared to 6.0 ± 1.0 min in the control group (p < 0.05). The cost of materials used was significantly higher in the study group, with an average of 300 ± 25 USD, compared to 200 ± 20 USD in the control group (p < 0.05). Additionally, the two groups showed no significant difference in the need for experience and surveillance (p > 0.05). Significantly lower visual analog scale (VAS) scores were found one day after the surgery in patients given dexmedetomidine plus midazolam than those anesthetized administered with dexmedetomidine only, suggesting an enhanced pain mitigation effect after incorporating midazolam for anesthetic induction. Patients treated with dexmedetomidine plus midazolam presented with a more stable hemodynamic status than those treated with dexmedetomidine only, as evidenced by the significantly lower variability of mean arterial pressure (MAP), oxygen saturation (SpO2), and heart rate (HR). Co-administration of dexmedetomidine plus midazolam for lobectomy anesthesia resulted in significantly lower serum cortisol (Cor) and norepinephrine (NE) concentrations in patients at anesthesia recovery than dexmedetomidine alone. However, this difference was not observed one day postoperatively. There was no statistically significant difference in the incidence of adverse reactions between the two groups. CONCLUSION The combination of midazolam with dexmedetomidine anesthesia in lobectomy improves the intraoperative hemodynamic status of elderly patients with NSCLC and mitigates their stress response. However, further research is required to explore the underlying mechanisms.
Collapse
Affiliation(s)
- Yanjun Zhao
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Dongjiao An
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Liang Bi
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Yang F, Liu J, Xu M, Peng B. Acquired multiple EGFR mutations‑mediated resistance to a third‑generation tyrosine kinase inhibitor in a patient with lung adenocarcinoma who responded to afatinib: A case report and literature review. Oncol Lett 2025; 29:81. [PMID: 39655272 PMCID: PMC11626421 DOI: 10.3892/ol.2024.14827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/02/2024] [Indexed: 12/12/2024] Open
Abstract
For patients with advanced non-small cell lung cancer (NSCLC) that have epidermal growth factor receptor (EGFR) mutations, EGFR tyrosine kinase inhibitors (TKIs) are the standard treatment and have significant clinical benefits. Third-generation TKIs, such as osimertinib, almonertinib and furmonertinib, are effective for the treatment of NSCLC that is EGFR-sensitizing mutation-positive and T790M-positive. Despite the efficacy of third-generation TKIs, patients inevitably develop resistance and the resistance mechanisms are heterogeneous. Second-generation inhibitors, such as afatinib, may be crucial in treating diseases that have developed resistance to first- or third-generation inhibitors. However, the clinical effect of afatinib in patients with acquired multiple EGFR mutations is not well defined. To the best of our knowledge, the present report describes the first case of a patient with lung adenocarcinoma who had multiple co-existing EGFR resistance mutations, including EGFR L718Q, EGFR C797S, EGFR C797G, EGFR L792H, EGFR V802F and EGFR V689L. These mutations conferred resistance to almonertinib, whilst maintaining sensitivity to afatinib.
Collapse
Affiliation(s)
- Fang Yang
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518036, P.R. China
| | - Jingjing Liu
- Department of Thoracic Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, P.R. China
| | - Mingming Xu
- Department of Thoracic Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, P.R. China
| | - Bin Peng
- Department of Thoracic Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, P.R. China
| |
Collapse
|
3
|
Zhuang J, Jiang H, Lou J, Zhang Y. TtAgo-coupled-multiplex-digtal-RPA-CRISPR/Cas12a (TCMDC) for EGFR mutations detection. Talanta 2025; 283:127162. [PMID: 39522277 DOI: 10.1016/j.talanta.2024.127162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/30/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
Epidermal Growth Factor Receptor (EGFR) is an important target for the early evaluation, treatment, and postoperative follow-up in non-small cell lung cancer (NSCLC). Current detection technologies suffer from extended detection time and high rate of false positive amplification. Therefore, the development of rapid, highly sensitive and specific detection methods is of great significance for improving the diagnosis and treatment of lung cancer. In this study, we proposed a fast and sensitive detection method termed Thermus thermophilus Argonaute (Ttago)-Coupled-Multiplex-digital-recombinase polymerase amplification (RPA)-Clustered regularly interspaced short palindromic repeats (CRISPR)/Cas12a (TCMDC) detection method, integrating EGFR mutation template enrichment. Based on the cleavage principle of TtAgo, the wild type (WT) template was enriched under the action of double-guide DNA. Two CRISPR RNAs, not restricted by protospacer adjacent motif (PAM) sites, were introduced to target EGFR genes. By combining RPA with CRISPR-Cas12a, we established a single-pot, ultra-sensitive (1 copy, 0.1 %), and visually detectable method for EGFR detection. We further verified the feasibility of this approach using clinical serum samples from lung cancer patients, achieving rapid (within 1 h) and visual detection of EGFR, thereby presenting a promising clinical tool for the detection of lung cancer.
Collapse
Affiliation(s)
- Jianjian Zhuang
- Department of Clinical pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Westlake University School of Medicine, Hangzhou, Zhejiang, 310006, China.
| | - Hong Jiang
- Department of Clinical pharmacology, Affiliated Hangzhou First People's Hospital, Cancer Center, Westlake University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Jiang Lou
- Department of Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Westlake University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Yu Zhang
- Department of Clinical pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Westlake University School of Medicine, Hangzhou, Zhejiang, 310006, China
| |
Collapse
|
4
|
Zhang X, Wang R, Zhang X, Yang Y, Tian R. Ferroptosis related CPT1A and GDF15 gene polymorphisms are risk factors for lung adenocarcinoma: A case-control study. Gene 2025; 933:149002. [PMID: 39401734 DOI: 10.1016/j.gene.2024.149002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/28/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Ferroptosis is not only a consequence of inflammation, but also a dynamic process. Recent bioinformatics analysis suggests that ferroptosis related genes might be associated with lung adenocarcinoma (LUAD). CPT1A and GDF15 are critical for the process of ferroptosis and development of inflammation; however, little study focused on the mutation level of these genes in patients with LUAD. METHODS The candidate SNPs in CPT1A and GDF15 were genotyped in 320 pairs of LUAD patients and controls using Mass ARRAY platform. Moreover, the different expression of CPT1A and GDF15 in LUAD cases and healthy controls were validated by qRT-PCR and ELISA. RESULTS The rs80356779 G > A, rs3019594 C > T, rs888663 T > G and rs4808793 G > C all exhibited an increased risk of the disease (p < 0.05). Moreover, the rs80356779-GA, rs3019594-TT, rs888663-TG and rs4808793-CC genotypes were all related to different levels of increase in LUAD risk (p < 0.05). Genetic model results showed that rs80356779 G > A, rs888663 T > G and rs4808793 G > C were associated with LUAD susceptibility under dominant and additive models (p < 0.05), while rs3019594 C > T was correlated with an elevated risk of the disease in all three models (p < 0.05). Additionally, patients with rs80356779 G > A and rs3019594 C > T exhibited lower expression and serum concentration of CPT1A compared with wile types, and patients with rs888663 T > G and rs4808793 G > C exhibited higher serum and expression level of GDF15. CONCLUSION The results provided new clues for the role of ferroptosis in LUAD and new potential targets for screening of susceptible population.
Collapse
Affiliation(s)
- Xing Zhang
- Respiratory department, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Science/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Rong Wang
- Respiratory department, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Science/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Xia Zhang
- Respiratory department, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Science/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Yanli Yang
- Respiratory department, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Science/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Ruifen Tian
- Respiratory department, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Science/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
5
|
Huang X, Yu G, Jiang X, Shen F, Wang D, Wu S, Mi Y. ITGB4/GNB5 axis promotes M2 macrophage reprogramming in NSCLC metastasis. Int Immunopharmacol 2025; 144:113564. [PMID: 39577216 DOI: 10.1016/j.intimp.2024.113564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/21/2024] [Accepted: 10/31/2024] [Indexed: 11/24/2024]
Abstract
OBJECTIVE Metastasis of non-small cell lung cancer (NSCLC) is a leading cause of high mortality. In recent years, the role of M2 macrophages in promoting tumor metastasis within the tumor microenvironment has garnered increasing attention. This study aims to investigate the role and potential mechanisms of the ITGB4/GNB5 axis in regulating M2 macrophage reprogramming and influencing NSCLC metastasis. METHODS This study first used single-cell sequencing technology to reveal the diverse subpopulation structure of NSCLC tumor tissues. Data analysis then identified the correlation between M2 macrophages and the malignant phenotype of NSCLC. Flow cytometry and immunohistochemistry were used to detect changes in M2 macrophages in NSCLC tissues. The impact of the ITGB4/GNB5 axis on M2 macrophage function was assessed through RNA sequencing and proteomic analysis. Finally, in vitro cell experiments and in vivo mouse models were used to validate the function and regulatory mechanisms of this axis. RESULTS Our study found diverse cellular subpopulations in NSCLC tumor tissues, with M2 macrophages closely associated with the malignant phenotype of NSCLC. We identified ITGB4 as a characteristic gene of NSCLC and predicted GNB5 as an interacting gene through database analysis. Activation of the ITGB4/GNB5 axis was shown to enhance M2 macrophage polarization, promoting their accumulation in the tumor microenvironment. This change further facilitated NSCLC invasion and metastasis by modulating related cytokines and signaling pathways. Animal experiments demonstrated that inhibition of the ITGB4/GNB5 axis significantly reduced tumor growth and metastasis. CONCLUSION The ITGB4/GNB5 axis reshapes the TME by promoting M2 macrophage polarization and functional enhancement, thereby facilitating tumor invasion and metastasis in NSCLC. This research provides new insights into the molecular mechanisms of NSCLC and offers potential molecular targets for future targeted therapies.
Collapse
Affiliation(s)
- Xiaofeng Huang
- Department of Cardiothoracic Surgery, Jiangyin Clinical College of Xuzhou Medical University, Jiangyin 214400, China
| | - Guiping Yu
- Department of Cardiothoracic Surgery, Jiangyin Clinical College of Xuzhou Medical University, Jiangyin 214400, China
| | - Xuewei Jiang
- Department of Cardiothoracic Surgery, Jiangyin Clinical College of Xuzhou Medical University, Jiangyin 214400, China
| | - Fei Shen
- Department of Cardiothoracic Surgery, Jiangyin Clinical College of Xuzhou Medical University, Jiangyin 214400, China
| | - Dengshu Wang
- Department of Cardiothoracic Surgery, Jiangyin Clinical College of Xuzhou Medical University, Jiangyin 214400, China
| | - Song Wu
- Department of Cardiothoracic Surgery, Jiangyin Clinical College of Xuzhou Medical University, Jiangyin 214400, China.
| | - Yedong Mi
- Department of Cardiothoracic Surgery, Jiangyin Clinical College of Xuzhou Medical University, Jiangyin 214400, China.
| |
Collapse
|
6
|
Lin F, Shen J, Li H, Liu L. β-carboline compound-10830733 suppresses the progression of non-small cell lung cancer by inhibiting the PI3K/Akt/GSK 3β signaling pathway. Eur J Pharmacol 2025; 986:177131. [PMID: 39566811 DOI: 10.1016/j.ejphar.2024.177131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024]
Abstract
Lung cancer is one of the most commonly diagnosed cancers worldwide, with non-small cell lung cancer (NSCLC) accounting for 80-85% of cases. To clarify the mechanisms underlying its onset and development, and to identify small molecule compounds that target related pathways effectively inhibiting tumor development and transformation. Small molecular compounds with a β-carboline nucleus exhibit a range of biological activities, with significant anti-tumor effects. A series of small molecule β-carboline compounds were synthesized and the dominant structure 1- (3-chlorophenyl) - 9H -pyridino - [3,4-b] indole - 3 -carboxylic acid methyl ester (10830733) was initially screened out. However, the effect of 10830733 on NSCLC is unclear. In this study, we investigated the anti-NSCLC activity of 10830733 and explored its potential mechanisms of action. First, we found that 10830733 decreased proliferation and invasion and promoted apoptosis, as well as S and G2 phase cell cycle arrest in NSCLC cells. Furthermore, network pharmacological analysis and Western blot confirmed that 10830733 inhibits the PI3K/Akt/GSK 3β pathway, and that the PI3K inhibitor LY294002 enhances the effects of 10830733 on proliferation, invasion, apoptosis, S and G2 phase arrest, and the expression of PI3K/Akt/GSK 3β related proteins. In conclusion, our data demonstrate that 10830733 reduces proliferation and invasion, promotes S and G2 phase arrest and apoptotic cell death in NSCLC cells by suppressing the PI3K/Akt/GSK 3β signaling pathway, suggesting that 10830733 could be a promising new candidate for NSCLC therapy.
Collapse
Affiliation(s)
- Fangrui Lin
- Department of Basic Medicine, Hebei University, Baoding, 071000, Heibei, China
| | - Junmin Shen
- Department of Basic Medicine, Hebei University, Baoding, 071000, Heibei, China
| | - Hangyu Li
- Department of Basic Medicine, Hebei University, Baoding, 071000, Heibei, China
| | - Li Liu
- Department of Basic Medicine, Hebei University, Baoding, 071000, Heibei, China.
| |
Collapse
|
7
|
Lv LL, Zhai JW, Wu JJ, Fan GQ, Zhang YX, Shen Y, Qu QX, Chen C. High CD38 expression defines a mitochondrial function-adapted CD8 + T cell subset with implications for lung cancer immunotherapy. Cancer Immunol Immunother 2025; 74:49. [PMID: 39751818 PMCID: PMC11699171 DOI: 10.1007/s00262-024-03881-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 11/03/2024] [Indexed: 01/04/2025]
Abstract
Despite identifying specific CD8+ T cell subsets associated with immunotherapy resistance, the molecular pathways driving this process remain elusive. Given the potential role of CD38 in regulating CD8+ T cell function, we aimed to investigate the accumulation of CD38+CD8+ T cells in lung cancer and explore its role in immunotherapy resistance. Phenotypic analysis of tumoral CD8+ T cells from both lung cancer patients and immunotherapy-resistant preclinical models revealed that CD38-expressing CD8+ T cells consist of CD38hi and CD38int subsets. These cells exhibited higher expression of exhaustion markers and displayed dysregulated mitochondrial bioenergetics. Notably, increased levels of CD38hiCD8+ T cells in the peripheral, but not central, tumor microenvironment were associated with a favorable response to anti-PD-1 therapy in non-small-cell lung cancer and correlated with the depth of clinical regression. This was evidenced by the greater depletion of CD38hiCD8+ T cells in patients with higher regional CD38hiCD8+ T cell infiltration. In immune checkpoint blockade (ICB)-resistant murine lung cancer models, PD-L1 mAbs alone failed to effectively reduce CD38hiCD8+ T cell levels. Notably, combination therapy with PD-L1 mAbs and EGCG selectively restricted CD38hiCD8+ T cell infiltration and enhanced IFN-γ production, significantly improving survival in this carcinoma model. The restoration of immunotherapy sensitivity was linked to improved mitochondrial function in CD38hiCD8+ T cells, which was validated by the established relationship between IFN-γ production and mitochondrial metabolism. Collectively, our data highlight the role of CD38-coupled mitochondrial dysfunction in promoting CD8+ T cell exhaustion and intrinsic resistance to ICB therapy, thereby offering a rationale for targeting CD38 to enhance the therapeutic efficacy of PD-1 blockade in lung cancer.
Collapse
Affiliation(s)
- Lei-Lei Lv
- Department of Respiratory and Critical Medicine, the First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, 215006, China
| | - Jia-Wei Zhai
- Department of Respiratory and Critical Medicine, the First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, 215006, China
- Respiratory Department, Xuzhou Central Hospital, 199 Jiefangnan Road, Xuzhou, 221000, China
| | - Jia-Juan Wu
- Clinical Immunology Institute, the First Affiliated Hospital of Soochow University, 178 Ganjiang Road, Suzhou, 215006, China
| | - Gui-Qin Fan
- Respiratory Department, Taicang Traditional Chinese Medicine Hospital, 140 Renmin South Road, Taicang, 215400, China
| | - Yao-Xin Zhang
- Department of Respiratory and Critical Medicine, the First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, 215006, China
| | - Yu Shen
- Clinical Immunology Institute, the First Affiliated Hospital of Soochow University, 178 Ganjiang Road, Suzhou, 215006, China
| | - Qiu-Xia Qu
- Clinical Immunology Institute, the First Affiliated Hospital of Soochow University, 178 Ganjiang Road, Suzhou, 215006, China.
| | - Cheng Chen
- Department of Respiratory and Critical Medicine, the First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, 215006, China.
| |
Collapse
|
8
|
Yin G, Liu X, Yu X, Tan S, Liu F. Analysis of ICIs alone or in combination rechallenged outcomes after progression from first-line ICIs plus chemotherapy in patients with advanced non-small cell lung cancer. Sci Rep 2025; 15:30. [PMID: 39747923 PMCID: PMC11696066 DOI: 10.1038/s41598-024-83947-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) plus chemotherapy have become the standard of care for first-line treatment of advanced non-small cell lung cancer (NSCLC) with EGFR/ALK negative. However, there is no clear second-line treatment option after first-line treatment failure. To investigate the efficacy and safety of ICIs alone or in combination rechallenge treatment after first-line ICIs plus chemotherapy progression in advanced NSCLC. We retrospectively analyzed the cases of patients who received ICIs alone or in combination rechallenge treatment after first-line ICIs plus chemotherapy progression in advanced NSCLC at Hunan Cancer Hospital between January 2020 and May 2024. We evaluated the effects of continued immunotherapy on patients' objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and adverse events after first-line treatment progression, and analyzed the relationship between outcomes and clinical characteristics. A total of 154 patients were included, with 146 patients developing resistance, 8 patients showing no progression. The ORR was 16.44%, the DCR was 68.49%, and the median PFS was 4.6 months. Patients treated with the new immune drug therapy had longer PFS than those treated with the original immunotherapy (5.0 months vs. 3.7 months, p = 0.0438). The PFS in patients receiving ICIs plus targeted therapy was significantly longer than that in patients who receiving ICIs alone, chemo-ICIs plus targeted therapy and ICIs plus chemotherapy (chemo-ICIs) (5.7 months vs. 3.6 months vs3.2 months vs. 2.9 months, p = 0.0086). Multivariate analysis showed that treatment regimen was a risk factor for immune rechallenge PFS, but there was no statistical correlation between gender, age, smoking history, pathological type, intermittent treatment or first-line drug resistance and immune rechallenge PFS. Our findings suggest that selecting ICIs plus targeted therapy may improve PFS in patients with advanced NSCLC after first-line chemo-ICIs progression. while replacement with new BSAb/PD-1 may be more beneficial to patients. However, there is a lack of large sample randomized controlled studies and evidence-based medical evidence, and more clinical studies are needed to further confirm.
Collapse
Affiliation(s)
- Guisen Yin
- Department of Pharmacy, Yantai Hospital of Traditional Chinese Medicine, Yantai, 264000, Shandong, China
| | - Xin Liu
- Department of Oncology, Yantai Hospital of Traditional Chinese Medicine, Yantai, 264000, Shandong, China
| | - Xiangtao Yu
- Department of Pharmacy, Yantai Hospital of Traditional Chinese Medicine, Yantai, 264000, Shandong, China
| | - Song Tan
- Department of Oncology, Yantai Hospital of Traditional Chinese Medicine, Yantai, 264000, Shandong, China
| | - Fen Liu
- Department of Pharmacy, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
9
|
Zhang W, Song L, Zhou Y, Sun J, Li C, Han B, Chang J, Han B, Wang T. Study on the inhibition of non-small cell lung cancer mediated by chitosan-based gene carrier delivering STAT3-shRNA. Int J Biol Macromol 2025; 284:138211. [PMID: 39617245 DOI: 10.1016/j.ijbiomac.2024.138211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/10/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Systemic chemotherapy and radiotherapy often yield poor effect in the postoperative treatment of non-small cell lung cancer (NSCLC) and induce drug resistance. Herein, we proposed a targeted therapeutic approach utilizing gene carrier-mediated specific shRNA method. Firstly, the targeted short hairpin shRNA sequence, designed based on the STAT3 gene sequence, was inserted into the eukaryotic expression vector pGPU6/GFP/Neo to form the recombinant plasmid STAT3-shRNA. Next, a novel gene carrier, Vitamin E Succinate-Chitosan-Histidine (VES-CTS-His, VCH), was synthesized through an acylation reaction. The VCH was combined with pGPU6/GFP/Neo STAT3-shRNA recombinant plasmid by electrostatic interactions to form stable particles. VCH/pDNA, with typical nanoscale dimensions, could accumulate in tumor tissues through the EPR effect and enter tumor cells via endocytosis. VCH exhibited good pH responsiveness and could dissociate in the acidic microenvironment of tumors, thereby releasing the plasmids. Subsequently, the plasmids could downregulate STAT3 expression through RNAi effect. Inhibiting or blocking the expression of the STAT3 gene could significantly enhance the apoptotic induction and growth inhibition effects on NSCLC cells through the PI3K and mTOR signaling pathways, thereby achieving the goal of tumor treatment. This study provides a novel method for the construction of novel non-viral gene carriers and clinical gene-targeted therapy for NSCLC.
Collapse
Affiliation(s)
- Wangwang Zhang
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Leyang Song
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Yi Zhou
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Jishang Sun
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Cuiyao Li
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Baoqin Han
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, PR China
| | - Jing Chang
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, PR China.
| | - Baosan Han
- Department of Breast Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| | - Ting Wang
- Department of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China.
| |
Collapse
|
10
|
Zhao Y, Xiong S, Ren Q, Wang J, Li M, Yang L, Wu D, Tang K, Pan X, Chen F, Wang W, Jin S, Liu X, Lin G, Yao W, Cai L, Yang Y, Liu J, Wu J, Fu W, Sun K, Li F, Cheng B, Zhan S, Wang H, Yu Z, Liu X, Zhong R, Wang H, He P, Zheng Y, Liang P, Chen L, Hou T, Huang J, He B, Song J, Wu L, Hu C, He J, Yao J, Liang W. Deep learning using histological images for gene mutation prediction in lung cancer: a multicentre retrospective study. Lancet Oncol 2025; 26:136-146. [PMID: 39653054 DOI: 10.1016/s1470-2045(24)00599-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Accurate detection of driver gene mutations is crucial for treatment planning and predicting prognosis for patients with lung cancer. Conventional genomic testing requires high-quality tissue samples and is time-consuming and resource-consuming, and as a result, is not available for most patients, especially those in low-resource settings. We aimed to develop an annotation-free Deep learning-enabled artificial intelligence method to predict GEne Mutations (DeepGEM) from routinely acquired histological slides. METHODS In this multicentre retrospective study, we collected data for patients with lung cancer who had a biopsy and multigene next-generation sequencing done at 16 hospitals in China (with no restrictions on age, sex, or histology type), to form a large multicentre dataset comprising paired pathological image and multiple gene mutation information. We also included patients from The Cancer Genome Atlas (TCGA) publicly available dataset. Our developed model is an instance-level and bag-level co-supervised multiple instance learning method with label disambiguation design. We trained and initially tested the DeepGEM model on the internal dataset (patients from the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China), and further evaluated it on the external dataset (patients from the remaining 15 centres) and the public TCGA dataset. Additionally, a dataset of patients from the same medical centre as the internal dataset, but without overlap, was used to evaluate the model's generalisation ability to biopsy samples from lymph node metastases. The primary objective was the performance of the DeepGEM model in predicting gene mutations (area under the curve [AUC] and accuracy) in the four prespecified groups (ie, the hold-out internal test set, multicentre external test set, TCGA set, and lymph node metastases set). FINDINGS Assessable pathological images and multigene testing information were available for 3697 patients who had biopsy and multigene next-generation sequencing done between Jan 1, 2018, and March 31, 2022, at the 16 centres. We excluded 60 patients with low-quality images. We included 3767 images from 3637 consecutive patients (1978 [54·4%] men, 1514 [41·6%] women, 145 [4·0%] unknown; median age 60 years [IQR 52-67]), with 1716 patients in the internal dataset, 1718 patients in the external dataset, and 203 patients in the lymph node metastases dataset. The DeepGEM model showed robust performance in the internal dataset: for excisional biopsy samples, AUC values for gene mutation prediction ranged from 0·90 (95% CI 0·77-1·00) to 0·97 (0·93-1·00) and accuracy values ranged from 0·91 (0·85-0·98) to 0·97 (0·93-1·00); for aspiration biopsy samples, AUC values ranged from 0·85 (0·80-0·91) to 0·95 (0·86-1·00) and accuracy values ranged from 0·79 (0·74-0·85) to 0·99 (0·98-1·00). In the multicentre external dataset, for excisional biopsy samples, AUC values ranged from 0·80 (95% CI 0·75-0·85) to 0·91 (0·88-1·00) and accuracy values ranged from 0·79 (0·76-0·82) to 0·95 (0·93-0·96); for aspiration biopsy samples, AUC values ranged from 0·76 (0·70-0·83) to 0·87 (0·80-0·94) and accuracy values ranged from 0·76 (0·74-0·79) to 0·97 (0·96-0·98). The model also showed strong performance on the TCGA dataset (473 patients; 535 slides; AUC values ranged from 0·82 [95% CI 0·71-0·93] to 0·96 [0·91-1·00], accuracy values ranged from 0·79 [0·70-0·88] to 0·95 [0·90-1·00]). The DeepGEM model, trained on primary region biopsy samples, could be generalised to biopsy samples from lymph node metastases, with AUC values of 0·91 (95% CI 0·88-0·94) for EGFR and 0·88 (0·82-0·93) for KRAS and accuracy values of 0·85 (0·80-0·88) for EGFR and 0·95 (0·92-0·96) for KRAS and showed potential for prognostic prediction of targeted therapy. The model generated spatial gene mutation maps, indicating gene mutation spatial distribution. INTERPRETATION We developed an AI-based method that can provide an accurate, timely, and economical prediction of gene mutation and mutation spatial distribution. The method showed substantial potential as an assistive tool for guiding the clinical treatment of patients with lung cancer. FUNDING National Natural Science Foundation of China, the Science and Technology Planning Project of Guangzhou, and the National Key Research and Development Program of China. TRANSLATION For the Chinese translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China; AI Lab, Tencent, Shenzhen, China
| | - Shan Xiong
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China; Department of Thoracic Oncology and Surgery, Hengqin Hospital, The First Affiliated Hospital of Guangzhou Medical University, Hengqin, China
| | - Qin Ren
- AI Lab, Tencent, Shenzhen, China
| | - Jun Wang
- AI Lab, Tencent, Shenzhen, China
| | - Min Li
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Yang
- Department of Thoracic Surgery, Shenzhen People's Hospital, 2nd Clinical Medical College of Jinan University, Shenzhen, China
| | - Di Wu
- Department of Respiratory Medicine, Shenzhen People's Hospital, 2nd Clinical Medical College of Jinan University, Shenzhen, China
| | - Kejing Tang
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaojie Pan
- Department of Thoracic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Fengxia Chen
- Department of Thoracic Surgery, Hainan General Hospital, Haikou, China
| | - Wenxiang Wang
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Shi Jin
- Department of Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Xianling Liu
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Gen Lin
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Wenxiu Yao
- Department of Oncology, University of Electronic Science and Technology of China, Sichuan Cancer Hospital and Institute & Cancer, The Second People's Hospital of Sichuan Province, Chengdu, China
| | - Linbo Cai
- Department of Oncology, Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Yi Yang
- Department of Thoracic Surgery, Chengdu Third People's Hospital, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Jixian Liu
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Jingxun Wu
- Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Wenfan Fu
- Department of Chest Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Kai Sun
- AI Lab, Tencent, Shenzhen, China
| | - Feng Li
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Bo Cheng
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Shuting Zhan
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Haixuan Wang
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Ziwen Yu
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Xiwen Liu
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ran Zhong
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Huiting Wang
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Ping He
- Department of Pathology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongmei Zheng
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Peng Liang
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | | | - Ting Hou
- Burning Rock Biotech, Guangzhou, China
| | | | - Bing He
- AI Lab, Tencent, Shenzhen, China
| | - Jiangning Song
- Biomedicine Discovery Institute and Monash Data Futures Institute, Monash University, Melbourne, VIC, Australia
| | - Lin Wu
- Department of Thoracic Medical Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Chengping Hu
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
| | - Jianxing He
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | | | - Wenhua Liang
- Department of Thoracic Oncology and Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China; Department of Thoracic Oncology and Surgery, Hengqin Hospital, The First Affiliated Hospital of Guangzhou Medical University, Hengqin, China.
| |
Collapse
|
11
|
Chen C, Luo Y, Hou Q, Qiu J, Yuan S, Deng K. A vision transformer-based deep transfer learning nomogram for predicting lymph node metastasis in lung adenocarcinoma. Med Phys 2025; 52:375-387. [PMID: 39341208 DOI: 10.1002/mp.17414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 08/06/2024] [Accepted: 08/12/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Lymph node metastasis (LNM) plays a crucial role in the management of lung cancer; however, the ability of chest computed tomography (CT) imaging to detect LNM status is limited. PURPOSE This study aimed to develop and validate a vision transformer-based deep transfer learning nomogram for predicting LNM in lung adenocarcinoma patients using preoperative unenhanced chest CT imaging. METHODS This study included 528 patients with lung adenocarcinoma who were randomly divided into training and validation cohorts at a 7:3 ratio. The pretrained vision transformer (ViT) was utilized to extract deep transfer learning (DTL) feature, and logistic regression was employed to construct a ViT-based DTL model. Subsequently, the model was compared with six classical convolutional neural network (CNN) models. Finally, the ViT-based DTL signature was combined with independent clinical predictors to construct a ViT-based deep transfer learning nomogram (DTLN). RESULTS The ViT-based DTL model showed good performance, with an area under the curve (AUC) of 0.821 (95% CI, 0.775-0.867) in the training cohort and 0.825 (95% CI, 0.758-0.891) in the validation cohort. The ViT-based DTL model demonstrated comparable performance to classical CNN models in predicting LNM, and the ViT-based DTL signature was then used to construct ViT-based DTLN with independent clinical predictors such as tumor maximum diameter, location, and density. The DTLN achieved the best predictive performance, with AUCs of 0.865 (95% CI, 0.827-0.903) and 0.894 (95% CI, 0845-0942), respectively, surpassing both the clinical factor model and the ViT-based DTL model (p < 0.001). CONCLUSION This study developed a new DTL model based on ViT to predict LNM status in lung adenocarcinoma patients and revealed that the performance of the ViT-based DTL model was comparable to that of classical CNN models, confirming that ViT was viable for deep learning tasks involving medical images. The ViT-based DTLN performed exceptionally well and can assist clinicians and radiologists in making accurate judgments and formulating appropriate treatment plans.
Collapse
Affiliation(s)
- Chuanyu Chen
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yi Luo
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Qiuyang Hou
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jun Qiu
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Shuya Yuan
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Kexue Deng
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
12
|
Xu S, Nie X, Li L, Bie ZX, Li YM, Zhang P, Qi J, Peng JZ, Li XG. Outcomes of First-Line Microwave Ablation of Treatment-Naive Epidermal Growth Factor Receptor-Mutated Advanced Lung Adenocarcinoma Treated with Tyrosine Kinase Inhibitors. J Vasc Interv Radiol 2025; 36:68-77.e3. [PMID: 39428057 DOI: 10.1016/j.jvir.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 10/03/2024] [Accepted: 10/09/2024] [Indexed: 10/22/2024] Open
Abstract
PURPOSE To investigate the outcomes of first-line image-guided microwave ablation (MWA) plus tyrosine kinase inhibitors (TKIs) in untreated epidermal growth factor receptor (EGFR)-mutant advanced lung adenocarcinoma (LUAD) and to compare with TKIs alone. MATERIALS AND METHODS This retrospective cohort study included patients between December 2015 and December 2021 and was divided into 2 groups (Group A: first-line MWA+TKIs; Group B: TKIs alone). Progression-free survival (PFS) was the primary end point, whereas overall survival (OS) was the secondary end point and were compared via the Kaplan-Meier methods. Univariate and multivariate analyses were used to investigate the predictors of PFS and OS. Propensity score matching (1:1 ratio) was applied between Group B and the subgroup of complete ablation in Group A. RESULTS A total of 117 patients were included (Group A: n = 43; Group B: n = 74). In a mean follow-up of 47.0 months (SD ± 19.4), Group A had significantly longer median PFS (19.0 vs 10.0 months; P < .001) and OS (41.0 vs 25.0 months; P = .044) than Group B. Predictors of PFS included first-line MWA (P < .001) and tumor stage (P = .020), while that of OS included first-line MWA (P = 0.039), tumor stage (P = 0.014), and usage of third-generation TKIs (P = 0.001). There were 23 pairs of patients obtained after propensity score matching (Group A1: complete ablation+TKIs; Group B1: TKIs alone). Group A1 had significantly longer median PFS (24.0 vs 10.0 months; P < .001) and OS (48.0 vs 24.0 months; P = .012) than Group B1. CONCLUSIONS First-line MWA significantly improved the outcomes of patients with untreated EGFR-mutant advanced LUAD treated with TKIs. Complete ablation predicted a better prognosis.
Collapse
Affiliation(s)
- Sheng Xu
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xin Nie
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Lin Li
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhi-Xin Bie
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuan-Ming Li
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Ping Zhang
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Qi
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jin-Zhao Peng
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao-Guang Li
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
13
|
Wang J, Guo C, Wang J, Zhang X, Qi J, Huang X, Hu Z, Wang H, Hong B. Tumor Mutation Signature Reveals the Risk Factors of Lung Adenocarcinoma with EGFR or KRAS Mutation. Cancer Control 2025; 32:10732748241307363. [PMID: 39760242 DOI: 10.1177/10732748241307363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Abstract
INTRODUCTION EGFR and KRAS mutations are frequently detected in lung adenocarcinoma (LUAD). Tumor mutational signature (TMS) determination is an approach to identify somatic mutational patterns associated with pathogenic factors. In this study, through the analysis of TMS, the underlying pathogenic factors of LUAD with EGFR and KRAS mutations were traced. METHODS This was a retrospective study. TMS of LUAD with KRAS and EGFR mutations from the TCGA, OncoSG, and MSK datasets was determined by two bioinformatics tools, namely the "MutationalPatterns" and "FitMS" packages. Elevated microsatellite alterations at selected tetranucleotide repeats (EMAST) of LUAD clinical specimens was analyzed using capillary electrophoresis. RESULTS In LUAD with KRAS mutations, TMS analysis indicated that the smoking-related SBS4 signature was enriched. For LUAD with EGFR L858R mutation, the smoking-related SBS4 signature was enriched in the Western population from the TCGA database; however, the smoking-related SBS4 signature was not obvious in Asian LUAD patients. LUAD with EGFR exon19 deletion (19Del) exhibited stronger SBS15 signature, which was related to defective DNA mismatch repair. Capillary electrophoresis analysis showed that an EMAST locus was frequently instable in LUAD with EGFR 19Del. Different from the Western population, Asian LUAD patients with EGFR mutations exhibited the enrichment of SBS1, SBS2, and SBS13 signatures, which were associated with the endogenous mutation process of cytidine deamination. CONCLUSIONS TMS analysis reveals that smoking is associated with LUAD with KRAS mutations. Defective DNA mismatch repair and endogenous cytidine deamination are associated with LUAD with EGFR mutations, especially for the EGFR 19Del. The endogenous mutational process is stronger in Asian LUAD patients than Western LUAD patients.
Collapse
Affiliation(s)
- Jialiang Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Chang Guo
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Jiexiao Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Xiaopeng Zhang
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Jian Qi
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Xiang Huang
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Zongtao Hu
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Hongzhi Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Bo Hong
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| |
Collapse
|
14
|
Lin QX, Song WW, Xie WX, Deng YT, Gong YN, Liu YR, Tian Y, Zhao WY, Tian L, Gu DN. Sequential treatment of anti-PD-L1 therapy prior to anti-VEGFR2 therapy contributes to more significant clinical benefits in non-small cell lung cancer. Neoplasia 2025; 59:101077. [PMID: 39561585 PMCID: PMC11617296 DOI: 10.1016/j.neo.2024.101077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024]
Abstract
OBJECTIVE Anti-angiogenic therapy and immune checkpoint blockade therapy are currently important treatments for non-small cell lung cancer. However, the combined use of the two therapies is controversial, and few studies have investigated the effects of different time sequences of the two therapies on treatment outcomes. METHODS The tumor-bearing mouse model was established and the mice were divided into four groups, including AA-ICB sequence group, ICB-AA sequence group, synchronization group and the control group. Immunohistochemistry was used to assess tumor microvessels and PD-L1 expression. Selected immune cell populations were evaluated using flow cytometry. Meta-analysis and clinical information were used to elucidate the clinical effects of administration sequence. RESULTS We found that anti-PD-L1 treatment followed by anti-VEGFR2 therapy exerts the best inhibitory effect on tumor growth. Different sequences of anti-angiogenic therapy and immune checkpoint blockade therapy resulted in different proportions of tumor microvessels and immune cell populations in the tumor microenvironment. We further revealed that the administration of anti-PD-L1 before anti-VEGFR brought more normalized tumor blood vessels and CD8+T cell infiltration and reduced immunosuppressive cells in the tumor microenvironment. Subsequent re-transplantation experiments confirmed the long-term benefits of this treatment strategy. The meta-analysis reinforced that immunotherapy prior to anti-angiogenic therapy or combination therapy have better therapeutic effects in advanced non-small cell lung cancer. CONCLUSION Our study demonstrated that the therapeutic effect of anti-angiogenic treatment after immune checkpoint therapy was superior to that of concurrent therapy, whereas anti-angiogenic therapy followed by immunotherapy did not bring more significant clinical benefits than independent monotherapy.
Collapse
Affiliation(s)
- Qiao-Xin Lin
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wen-Wen Song
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wen-Xia Xie
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yi-Ting Deng
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yan-Na Gong
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yi-Ru Liu
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yi Tian
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Ya Zhao
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Tian
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Dian-Na Gu
- Department of Medical Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
15
|
Zhang SX, Zhang CC, Hou RP, Cai XW, Liu J, Yu W, Zhang Q, Guo JD, Wang CL, Li HX, Zhu ZF, Fu XL, Feng W. Is postoperative radiotherapy effective in patients with completely resected pathologic stage IIIA(N2) non-small cell lung cancer? High-risk populations should consider it. Clin Transl Radiat Oncol 2025; 50:100889. [PMID: 39634198 PMCID: PMC11616551 DOI: 10.1016/j.ctro.2024.100889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/18/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024] Open
Abstract
Background and purpose We aimed to assess the benefits of postoperative radiotherapy (PORT) in completely resected patients with pathologic stage IIIA(N2) non-small cell lung cancer (NSCLC) with a high risk of locoregional recurrence (LRR). Materials and methods A prospective, randomized trial was conducted starting in July 2016 to explore the optimal timing of PORT in high-LRR-risk patients with completely resected IIIA(N2) NSCLC (NCT02974426). Patients were identified as high-LRR-risk patients via the prognostic index (PI) model and were randomly assigned to PORT-first or PORT-last treatment. To evaluate PORT for high-LRR-risk patients, all patients in this trial constituted the PORT cohort, whereas high-LRR-risk patients without PORT were selected from a retrospective cohort as the non-PORT cohort. Propensity score-matched (PSM) analyses were conducted to compare overall survival (OS), disease-free survival (DFS), locoregional recurrence-free survival (LRFS) and distant metastasis-free survival (DMFS). Results Between 2016 and 2022, 132 patients were included in the trial, with a median follow-up of 49.3 months. The 3-year OS rate was 83.2 %, and the 3-year DFS rate was 35.0 %. Among these patients, 122 patients (92 %) received planned PORT. For 132 intention-to-treat patients, PSM analysis with the non-PORT cohort (n = 307) resulted in 130 matched pairs. The results revealed that PORT improved LRFS (3-year LRFS, 77.6 % vs. 57.3 %; p = 0.00014), DFS (3-year DFS, 35.2 % vs. 28.6 %; p = 0.038), and OS (3-year OS, 83.0 % vs. 60.7 %; p = 0.00017), with no difference in DMFS (p = 0.17). Conclusion PORT could increase local control, DFS, and OS in high-LRR-risk patients with completely resected IIIA(N2) NSCLC. Future research should utilize multidimensional data to pinpoint more precise subgroups benefiting from PORT, with prospective trials validating these findings.
Collapse
Affiliation(s)
- Shu-Xian Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen-Chen Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Run-Ping Hou
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xu-Wei Cai
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Liu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Yu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qin Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin-Dong Guo
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chang-Lu Wang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong-Xuan Li
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng-Fei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiao-Long Fu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Feng
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Mao F, Hu Z, Shi R, Zhang H, Zhang Z, Li Y, Li X, Gao P, Li J, Liu M, Liu H, Chen J. Unravelling the prognostic and operative role of intratumoural microbiota in non-small cell lung cancer: Insights from 16S rRNA and RNA sequencing. Clin Transl Med 2025; 15:e70156. [PMID: 39754314 DOI: 10.1002/ctm2.70156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/07/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Complex interrelationships between the microbiota and cancer have been identified by several studies. However, despite delineating microbial composition in non-small cell lung cancer (NSCLC), key pathogenic microbiota and their underlying mechanisms remain unclear. METHODS We performed 16S rRNA V3-V4 amplicon and transcriptome sequencing on cancerous and adjacent normal tissue samples from 30 patients with NSCLC, from which clinical characteristics and prognosis outcomes were collected. We used 16S rRNA sequencing to dissect microbial composition and perform prognosis correlations, and in conjunction with transcriptome sequencing, we determined potential mechanisms underpinning significant microbiota actions. RESULTS In comparing different sample types, we identified more pronounced beta diversity disparity between NSCLC, lung squamous cell carcinoma (LUSC) and corresponding paired normal tissues. Concurrently, LUSC and lung adenocarcinoma exhibited distinct microbial composition traits at genus levels. Subsequently, four phyla, five classes, nine orders, 17 families and 36 genera were filtered out and were related to prognosis outcomes. Intriguingly, a protective microbial cluster was identified encompassing nine genera associated with delayed disease recurrence, with functional analyses suggested that these microbiota predominantly exerted metabolism-related functions. Additionally, a harmful microbial cluster (HMC) was identified, including three genera. In this HMC and subsequent prognosis model analyses, harmful intratumoural microbiota were potentially implicated in infection, inflammation and immune regulation. Crucially, we identified a microbial genus, Peptococcus, which was as an independent, detrimental NSCLC prognostic factor and potentially impacted prognosis outcomes via tumour necrosis factor (TNF) signalling. CONCLUSIONS We identified a substantial connection between intratumoural microbiota and NSCLC prognosis outcomes. Protective microbiota primarily exerted metabolic functions, whereas harmful microbiota were mainly implicated in infection, inflammation and immune modulation. Furthermore, Peptococcus may be significant in adverse NSCLC prognoses and serve as a potential biomarker for patient management and cancer screening. KEY POINTS Four phyla, five classes, nine orders, 17 families and 36 genera have been found associated with NSCLC prognosis. We identified a protective microbial cluster associated with delayed recurrence and a harmful microbial cluster related to shorter survival and earlier recurrence. We identified Peptococcus as an independent, detrimental prognostic factor for NSCLC, potentially impacting prognosis via TNF signalling.
Collapse
Affiliation(s)
- Fuling Mao
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Zixuan Hu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Ruifeng Shi
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Hongbing Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Zihe Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yongwen Li
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xuanguang Li
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Penghu Gao
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinhui Li
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Minghui Liu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongyu Liu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jun Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
17
|
Geng H, Xue Y, Yan B, Lu Z, Yang H, Li P, Zhou J. Network Pharmacology and Molecular Docking Study on the Mechanism of the Therapeutic Effect of Strychni Semen in NSCLC. Biol Proced Online 2024; 26:33. [PMID: 39736533 DOI: 10.1186/s12575-024-00259-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 12/18/2024] [Indexed: 01/01/2025] Open
Abstract
Strychni Semen, characterized by its bitter taste and warm properties, has been confirmed to possess anti-tumor properties. However, the molecular mechanism of Strychni Semen in treating non-small cell lung cancer (NSCLC) needs further study. This study aimed to explore the molecular mechanism of Strychni Semen in treating NSCLC based on network pharmacology and molecular docking. The active components and targets of Strychni Semen were retrieved from the TCMSP, supplemented by the HERB database and the related literature. NSCLC-related targets were retrieved from the GeneCards, OMIM and DisGenet databases. The intersection targets of Strychni Semen in treating NSCLC were obtained via an online platform. The Protein-Protein Interaction (PPI) network was subsequently constructed to deeply analyse the interrelationship of the intersection targets via the String database. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were carried out via the Metascape database. The interactive networks between Strychni Semen and NSCLC were constructed via Cytoscape 3.9.1. Molecular docking detected interactions between the key components and the core targets. The core targets were validated via GEO datasets. 21 active components and 67 targets were identified, with 47 associated with NSCLC. The key active components were Stigmasterol, IcarideA, 2-Hydroxymethylanthraquinone, (+)-catechin, (2R)-5,7-dihydroxy-2-(4-hydroxyphenyl)chroman-4-one, (S)-Stylopine, Brucine and Isobrucine. The core targets were PTGS2, NR3C1, ESR1, CASP3 and PRKACA. Molecular docking revealed that these compounds undergo strong binding affinity with the core genes. GEO database indicated that PTGS2 was the most promising core target. In addition, Strychni Semen's effects on NSCLC involved mainly the Calcium pathway, the Estrogen pathway, and the cGMP-PKG and cAMP pathways. This study visually demonstrated the mechanism of the therapeutic effect of Strychni Semen in NSCLC through multiple components, targets and pathways which provides a basis for clinical treatment and further experimental research.
Collapse
Affiliation(s)
- He Geng
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian Cancer Hospital, Huaian, Jiangsu, China
| | - Yujie Xue
- Department of Pathology, Affiliated Huaian NO. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Binghua Yan
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian Cancer Hospital, Huaian, Jiangsu, China
| | - Zhaoxue Lu
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian Cancer Hospital, Huaian, Jiangsu, China
| | - Hengjin Yang
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian Cancer Hospital, Huaian, Jiangsu, China
| | - Peng Li
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian Cancer Hospital, Huaian, Jiangsu, China.
| | - Jundong Zhou
- Department of Radiation Oncology, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, China.
- Suzhou Cancer Center Core Laboratory, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, China.
| |
Collapse
|
18
|
Ren J, Zhao S, Lai J. Role and mechanism of COL3A1 in regulating the growth, metastasis, and drug sensitivity in cisplatin-resistant non-small cell lung cancer cells. Cancer Biol Ther 2024; 25:2328382. [PMID: 38530094 DOI: 10.1080/15384047.2024.2328382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/05/2024] [Indexed: 03/27/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is among the most difficult malignancies to treat. Type III collagen (COL3A1) can affect the progression and chemoresistance development of NSCLC. We herein explored the mechanism that drives COL3A1 dysregulation in NSCLC. Potential RNA-binding proteins (RBPs) and transcription factors (TFs) that could bind to COL3A1 were searched by bioinformatics. mRNA expression was detected by quantitative PCR. Protein expression was evaluated using immunoblotting and immunohistochemistry. The effects of the variables were assessed by gauging cell growth, invasiveness, migratory capacity, apoptosis, and cisplatin (DDP) sensitivity. The direct YY1/COL3A1 relationship was confirmed by ChIP and luciferase reporter experiments. Xenograft experiments were done to examine COL3A1's function in DDP efficacy. COL3A1 showed enhanced expression in DDP-resistant NSCLC. In H460/DDP and A549/DDP cells, downregulation of COL3A1 exerted inhibitory functions in cell growth, invasiveness, and migration, as well as promoting effects on cell DDP sensitivity and apoptosis. Mechanistically, ELAV-like RNA binding protein 1 (ELAVL1) enhanced the mRNA stability and expression of COL3A1, and Yin Yang 1 (YY1) promoted the transcription and expression of COL3A1. Furthermore, upregulation of COL3A1 reversed ELAVL1 inhibition- or YY1 deficiency-mediated functions in DDP-resistant NSCLC cells. Additionally, COL3A1 downregulation enhanced the anti-tumor efficacy of DDP in vivo. Our investigation demonstrates that COL3A1 upregulation, induced by both RBP ELAVL1 and TF YY1, exerts important functions in phenotypes of NSCLC cells with DDP resistance, offering an innovative opportunity in the treatment of drug-resistant NSCLC.
Collapse
Affiliation(s)
- Jiankun Ren
- Nursing School, Hebi Polytechnic, Hebi City, China
| | - Songwei Zhao
- Nursing School, Hebi Polytechnic, Hebi City, China
| | - Junyu Lai
- Department of Cardiology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| |
Collapse
|
19
|
Li C, Liang L, Liang J, Tian C, Wang J, Liu Y, Hong X, Gu F, Zhang K, Hu Y, Liu L, Zeng Y. IQGAP3 activates Hedgehog signaling to confer stemness and metastasis via up-regulating GLI1 in lung cancer. Sci Rep 2024; 14:31327. [PMID: 39732803 DOI: 10.1038/s41598-024-82793-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/09/2024] [Indexed: 12/30/2024] Open
Abstract
Lung cancer ranks as the most prevalent malignant neoplasm worldwide, contributing significantly to cancer-related mortality. Stemness is a well-recognized factor underlying radiotherapy resistance, recurrence and metastasis in non-small-cell lung cancer (NSCLC) patients. Our prior investigations have established the role of IQ motif containing GTPase-activating protein 3 (IQGAP3) in mediating radiotherapy resistance in lung cancer, but its impact on lung cancer stemness remains unexplored. Our bioinformatics analysis results revealed a significant correlation between IQGAP3 and lung cancer stemness. Moreover, we found that IQGAP3 depletion in lung cancer cells resulted in reduced migration, invasion and sphere-forming capabilities. Through RNA sequencing, we identified GLI1 as a pivotal downstream effector of IQGAP3. The knockdown of IQGAP3 led to the downregulation of GLI1 mRNA and protein levels, which impeded the activation of the Hedgehog-GLI1 signaling pathway. Further, our results also indicated that GLI1 is the primary effector mediating IQGAP3's biological functions in lung cancer. These findings elucidate the role of IQGAP3 in promoting lung cancer stemness and metastasis through the Hedgehog pathway, facilitated by GLI1, highlighting the potential of IQGAP3 as a promising therapeutic target for lung cancer treatment.
Collapse
Affiliation(s)
- Chang Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Limei Liang
- Department of Respiratory and Critical Care Medicine, TongjiMedical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jinyan Liang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chen Tian
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Juanjuan Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yuting Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaohua Hong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Feifei Gu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kai Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yue Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Li Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Yulan Zeng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
20
|
Chen P, Cheng L, Zhao C, Tang Z, Wang H, Shi J, Li X, Zhou C. Machine learning identifies immune-based biomarkers that predict efficacy of anti-angiogenesis-based therapies in advanced lung cancer. Int Immunopharmacol 2024; 143:113588. [PMID: 39556888 DOI: 10.1016/j.intimp.2024.113588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/18/2024] [Accepted: 11/05/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND The anti-angiogenic drugs showed remarkable efficacy in the treatment of lung cancer. Nonetheless, the potential roles of the intra-tumoral immune cell abundances and peripheral blood immunological features in prognosis prediction of patients with advanced lung cancer receiving anti-angiogenesis-based therapies remain unknown. In this study, we aimed to develop an immune-based model for early identification of patients with advanced lung cancer who would benefit from anti-angiogenesis-based therapies. METHODS We assembled the real-world cohort of 1058 stage III-IV lung cancer patients receiving the anti-angiogenesis-based therapies. We comprehensively evaluated the tumor immune microenvironment characterizations (CD4, CD8, CD68, FOXP3, and PD-L1) by multiplex immunofluorescence (mIF), as well as calculated the systemic inflammatory index by flow cytometry and medical record review. Based on the light gradient boosting machine (LightGBM) algorithm, a machine-learning model with meaningful parameters was developed and validated in real-world populations. RESULTS In the first-line anti-angiogenic therapy plus chemotherapy cohort (n = 385), the intra-tumoral proportion of CD68 + Macrophages and several circulating inflammatory indexes were significantly related to drug response (p < 0.05). Further, neutrophil to lymphocyte ratio (NLR), monocyte to lymphocyte ratio (MLR), the systemic inflammation response index (SIRI), and myeloid to lymphoid ratio (M:L) were identified to construct the non-invasive prediction model with high predictive performance (AUC: 0.799 for treatment response and 0.7006-0.915 for progression-free survival (PFS)). Additionally, based on the unsupervised hierarchical clustering results, the circulating cluster 3 with the highest levels of NLR, MLR, SIRI, and M: L had the worst PFS with the first-line anti-angiogenic therapy plus chemotherapy compared to other circulating clusters (2.5 months, 95 % confidence interval 2.3-2.7 vs. 6.0-9.7 months, 95 % confidence interval 4.9-11.1, p < 0.01). The predictive power of the machine-learning model in PFS was also validated in the anti-angiogenic therapy plus immunotherapy cohort (n = 103), the anti-angiogenic monotherapy cohort (n = 284), and the second-line anti-angiogenic therapy plus chemotherapy cohort (n = 286). CONCLUSIONS Integrating pre-treatment circulating inflammatory biomarkers could non-invasively and early forecast clinical outcomes for anti-angiogenic response in lung cancer. The immune-based prognostic model is a promising tool to reflect systemic inflammatory status and predict clinical prognosis for anti-angiogenic treatment in patients with stage III-IV lung cancer.
Collapse
Affiliation(s)
- Peixin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; School of Medicine, Tongji University, Shanghai 2000922, China
| | - Lei Cheng
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Chao Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Zhuoran Tang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; School of Medicine, Tongji University, Shanghai 2000922, China
| | - Haowei Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; School of Medicine, Tongji University, Shanghai 2000922, China
| | - Jinpeng Shi
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; School of Medicine, Tongji University, Shanghai 2000922, China
| | - Xuefei Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China.
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; School of Medicine, Tongji University, Shanghai 2000922, China.
| |
Collapse
|
21
|
Sun T, Yang Z. TFAP2C-mediated transcriptional activation of STEAP3 promotes lung squamous cell carcinoma progression by regulating the β-catenin pathway. Biol Direct 2024; 19:135. [PMID: 39716275 DOI: 10.1186/s13062-024-00584-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024] Open
Abstract
Six-transmembrane epithelial antigen of prostate 3 (STEAP3) is associated with the progression of several human malignancies. However, its role in lung squamous cell carcinoma (LUSC) remains unclear. We measured STEAP3 expression in LUSC cell lines and tissues. LUSC cells with stable STEAP3 overexpression and knockdown were obtained through G418 selection. Multiple assays were used to evaluate the malignant phenotypes of LUSC cells and the activation of the β-catenin signaling. The potential transcriptional regulatory factors of STEAP3 were predicted using the JASPAR database, and the correlation between transcription factor AP-2 gamma (TFAP2C) and STEAP3 was analyzed through the GEPIA database. The study evaluated the regulatory relationship between a potential transcription factor and STEAP3 through ChIP and luciferase reporter assays. Additionally, rescue assays were utilized to ascertain whether TFAP2C serves as the upstream regulatory factor of STEAP3, contributing to LUSC progression. Finally, tumor growth and metastasis were evaluated in vivo. STEAP3 expression was notably higher in LUSC, and its overexpression was linked to a poor prognosis. Moreover, STEAP3 overexpression activated the β-catenin pathway, thereby accelerating cell proliferation and metastasis. Conversely, STEAP3 knockdown had an anti-tumor effect in LUSC. Additionally, TFAP2C bound directly to the STEAP3 promoter and positively regulate its expression in LUSC. The anti-tumor effects of TFAP2C knockdown were partially reversed by STEAP3 overexpression. The study indicates that the TFAP2C/STEAP3 axis may be a therapeutic target for LUSC treatment. This enhances our understanding of lung carcinogenesis.
Collapse
Affiliation(s)
- Tong Sun
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, Liaoning, P. R. China
| | - Zhiguang Yang
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, Liaoning, P. R. China.
| |
Collapse
|
22
|
Yan B, Yao X, Zhao Z, Sun T, Zhao Y, Zou W, Wu D, Chang H. Learning curve for double-port video-assisted thoracoscopic lung segmentectomy: a propensity score matching study. J Cardiothorac Surg 2024; 19:681. [PMID: 39716291 DOI: 10.1186/s13019-024-03180-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 12/01/2024] [Indexed: 12/25/2024] Open
Abstract
OBJECTIVE To determine the learning curve for double-port video-assisted thoracoscopic (VATS) lung segmentectomy performed by the same surgical team in our center. METHODS We retrospectively collected clinical data from 193 patients who underwent double-port video-assisted thoracoscopic lung segmentectomy from March 2017 to March 2023. The operative time (OT) was analyzed using the cumulative sum (CUSUM) method, and two stages of the learning curve were obtained. Propensity score matching (PSM) was performed for age, sex, underlying disease, and single-segment resection via radius matching. The OT, estimated amount of intraoperative blood loss, and other complications were analyzed. RESULTS We generated a graph of the CUSUM of the OT and found that the learning curve could be differentiated into two stages: the learning stage (1st to 95th surgery) and the proficiency stage (96th to 193rd surgery). Before PSM, there were significant differences in the OT, extent of lymph node station dissection, amount of drainage on the day of surgery, amount of drainage on the first postoperative day, estimated amount of intraoperative blood loss, and length of hospital stay after surgery. There were no significant differences in the average amount of drainage 3 days after surgery, postoperative tube time, or number of intraoperative revolutions. However, after PSM, there were significant differences in the OT, number of lymph node stations removed, amount of drainage on the day of surgery, and amount of drainage on the first postoperative day. There were no significant differences in the estimated amount of intraoperative blood loss, length of hospital stay after surgery, average amount of drainage for 3 days after surgery, postoperative tube time, or number of intraoperative revolutions. CONCLUSION In our center, the learning curve for double-port video-assisted thoracoscopic lung segmentectomy transitions from the learning stage to the proficiency stage when the number of surgical cases reaches 95. There were significant differences in the OT, number of lymph node stations removed, amount of drainage on the day of surgery, and amount of drainage on the first postoperative day.
Collapse
Affiliation(s)
- Bixin Yan
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, Heilongjiang, 150000, China
| | - Xiao Yao
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, Heilongjiang, 150000, China
| | - Zhihong Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, Heilongjiang, 150000, China
| | - Tianhao Sun
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, Heilongjiang, 150000, China
| | - Yiming Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, Heilongjiang, 150000, China
| | - Wenbiao Zou
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, Heilongjiang, 150000, China
| | - Duofu Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, Heilongjiang, 150000, China
| | - Hao Chang
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, Heilongjiang, 150000, China.
| |
Collapse
|
23
|
Wang M, Qin Y, Ai X, Liu X. RBM15-dependent m6A modification mediates progression of non-small cell lung cancer cells. Mol Med 2024; 30:267. [PMID: 39716068 DOI: 10.1186/s10020-024-01018-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 11/28/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the predominant form of lung cancer, contributing significantly to global health and economic challenges. This study elucidated the role of RBM15 in NSCLC progression through its involvement in m6A modifications. METHODS RBM15 levels in NSCLC tissues and cells were assessed via RT-qPCR and Western blotting. The impact of RBM15 knockdown on NSCLC proliferation, invasion, and migration was evaluated using CCK-8, colony formation, and Transwell assays. Expression levels of KLF1, TRIM13, and ANXA8 were determined by RT-qPCR and Western blot. m6A methylation levels were analyzed, while RIP and MeRIP assays were employed to explore the interaction between YTHDF1/YTHDF2/m6A and KLF1/TRIM13, as well as KLF1 binding to the ANXA8 promoter. The ubiquitination of ANXA8 was examined through ubiquitination assays. Xenograft and metastasis models were utilized to assess RBM15's role in vivo. RESULTS RBM15 was found to be overexpressed in NSCLC. Silencing RBM15 led to decreased cell proliferation, invasion, and migration of NSCLC cells. RBM15 upregulated KLF1 and downregulated TRIM13 via YTHDF1/YTHDF2, resulting in the promotion of ANXA8 expression. KLF1 overexpression or TRIM13 downregulation partially reversed the suppressive effects of RBM15 knockdown on NSCLC cell proliferation. ANXA8, upregulated in NSCLC, mitigated the inhibitory effects of RBM15 silencing on malignant behaviors. In vivo, RBM15 downregulation hindered NSCLC cell proliferation and metastasis by modulating the KLF1-TRIM13/ANXA8 axis. CONCLUSION RBM15-mediated m6A methylation enhances KLF1 expression and suppresses TRIM13 via YTHDF1/YTHDF2, thereby promoting ANXA8 and facilitating NSCLC progression. These findings provide novel insights and potential therapeutic targets for NSCLC treatment.
Collapse
Affiliation(s)
- Man Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Yujiao Qin
- Department of Respiratory Medicine, The First Affiliated Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Xiaoqi Ai
- Department of Respiratory Medicine, The First Affiliated Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Xiuhua Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, Jilin, China.
| |
Collapse
|
24
|
Huang Q, Ren H, Bao Z, Jiang Y, Luo Y, Yao L. The impact of neoadjuvant immunotherapy on the clinical efficacy of stage IIIA-N2 non-small cell lung cancer patients. Int Immunopharmacol 2024; 146:113809. [PMID: 39708486 DOI: 10.1016/j.intimp.2024.113809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/24/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024]
Abstract
OBJECTIVE To explore the impact of neoadjuvant immunotherapy on the clinical efficacy of stage IIIA-N2 non-small cell lung cancer (NSCLC) patients. METHODS The retrospective study was conducted on 120 patients with stage IIIA-N2 NSCLC admitted to our hospital during April 2020 to April 2022. The control group received local chemotherapy, while the combination group received neoadjuvant immunotherapy on the basis of chemotherapy. The treatment efficacy, immune function, PD-1 and PD-L1 (SP142) expression levels, and changes in inflammatory factors were compared between the two groups. Kaplan Meier survival curve was used to analyze the overall survival rate. RESULTS Total effective rate in the control group was 53.33 % (15.00 % recovery + 16.66 % significant effect + 21.66 % effective), and the combined group had a higher total effective rate of 85.00 % (41.66 % recovery + 23.33 % significant effective + 20.00 % effective) (P < 0.001). After intervention, the combination group had largely increased immune indicators levels, including CD3+, CD4+ and CD4+/CD8+ (P < 0.001), but reduced levels of CD8+, PD-1 and PD-L1 (SP142) than the control group (P < 0.001). After intervention, the levels of inflammatory factors in the combination group were also reduced than the control group (P < 0.001). Compared to the control group with an adverse reaction rate of 31.66 % (8.33 % gastrointestinal reaction + 11.66 % hair loss + 6.66 % proteinuria + 5.00 % diarrhea), the combined group had much lower adverse reaction rate of 11.66 % (1.66 % gastrointestinal reaction + 5.00 % hair loss + 3.33 % proteinuria + 1.66 % diarrhea) (P < 0.05). After 24 months of follow-up, the overall survival rate was 58.33 % (35/60) and 40.00 % (24/60) in the combination group and the control group, respectively. The Kaplan Meier survival curve analysis showed a statistically significant difference in overall survival rate between the two groups (P < 0.05). CONCLUSION Neoadjuvant immunotherapy had a more significant therapeutic effect on stage IIIA-N2 NSCLC patients by reducing immunosuppressive and inflammatory factors, improving immune function, and reducing the occurrence of adverse reactions.
Collapse
Affiliation(s)
- Qin Huang
- Department of Oncology, The Second People's Hospital of Banan District, Chongqing 400054, China.
| | - Hongbo Ren
- Department of Oncology, The Second People's Hospital of Banan District, Chongqing 400054, China
| | - Zhonghui Bao
- Department of Oncology, The Second People's Hospital of Banan District, Chongqing 400054, China
| | - Yi Jiang
- Department of Oncology, The Second People's Hospital of Banan District, Chongqing 400054, China
| | - Yan Luo
- Department of Oncology, The Second People's Hospital of Banan District, Chongqing 400054, China
| | - Lan Yao
- Department of Oncology, The Second People's Hospital of Banan District, Chongqing 400054, China
| |
Collapse
|
25
|
Zhou Q, Li D, Liang Y, Long Y, Liu Y. SEC14L2 regulates the transport of cholesterol in non-small cell lung cancer through SCARB1. Lipids Health Dis 2024; 23:407. [PMID: 39696431 DOI: 10.1186/s12944-024-02401-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Inhibiting cholesterol metabolism has shown great potential in non-small cell lung cancer (NSCLC). However, the regulatory mechanism of the lipid metabolism key factor Sect. 14-like lipid binding 2 (SEC14L2) in NSCLC remains unclear. This study investigates the effects of differentially expressed genes related to cholesterol metabolism on the development of NSCLC. METHODS Cox regression and survival analysis were performed to screen cholesterol metabolism-related genes and predict survival prognosis in NSCLC patients. The proliferation and migration of NSCLC cells were assessed by CCK-8, EdU, colony formation and wound-healing assay. Cholesterol depletion and rescue trials were used to evaluate the effect of SEC14L2 on cholesterol transport in NSCLC cells. IF and Co-IP were used to analyze the targeting relationship between SEC14L2 and scavenger receptor class B member 1 (SCARB1). RESULTS SEC14L2 was a key gene related to prognosis in NSCLC patients and was highly expressed in A549 and Calu-1 cells. Subsequent studies demonstrated that knockdown of SEC14L2 significantly reduced the proliferation and migration of NSCLC cells, resulting in inhibited tumor growth. Furthermore, both in vitro and in vivo experiments indicated that SEC14L2 regulated cholesterol uptake. Silencing SEC14L2 partially counteracted the promotion of cholesterol content by MβCD-chol in A549 and Calu-1 cells. We then verified that there was a protein interaction between SEC14L2 and SCARB1. CONCLUSION SEC14L2 promoted cholesterol uptake in NSCLC cells by up-regulating SCARB1 expression, thereby promoting NSCLC development.
Collapse
Affiliation(s)
- Qianhui Zhou
- Department of Respiratory and Critical Care Medicine, Zhuzhou Central Hospital, No.116, Changjiang South Road, Tianyuan District, Zhuzhou, 412000, Hunan, China
| | - Dianwu Li
- Department of Respiratory and Critical Care Medicine, Zhuzhou Central Hospital, No.116, Changjiang South Road, Tianyuan District, Zhuzhou, 412000, Hunan, China
| | - Yanchao Liang
- Department of Respiratory and Critical Care Medicine, Zhuzhou Central Hospital, No.116, Changjiang South Road, Tianyuan District, Zhuzhou, 412000, Hunan, China
| | - Yunzhu Long
- Department of Infectious Diseases, Zhuzhou Central Hospital, No.116, Changjiang South Road, Tianyuan District, Zhuzhou, 412000, Hunan, China.
| | - Yi Liu
- Department of Respiratory and Critical Care Medicine, Zhuzhou Central Hospital, No.116, Changjiang South Road, Tianyuan District, Zhuzhou, 412000, Hunan, China.
| |
Collapse
|
26
|
Sun K, Wang P. A good response to furmonertinib fourth-line treatment of an advanced lung adenocarcinoma patient with EGFR exon20in and PIK3CA mutation: a case report and literature review. Front Oncol 2024; 14:1467722. [PMID: 39743996 PMCID: PMC11688600 DOI: 10.3389/fonc.2024.1467722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Background Lung cancer, including small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC), is the most prevalent cancer globally and remains the leading cause of cancer-related mortality. Epidermal growth factor receptor (EGFR) mutations, frequently observed in female NSCLC patients, have revolutionized treatment strategies with the advent of tyrosine kinase inhibitors (TKIs). These therapies significantly improve survival and are considered the standard of care for patients harboring EGFR mutations. However, most patients eventually develop resistance to EGFR-TKIs, leading to disease progression. Resistance mechanisms are classified as either EGFR-dependent or EGFR-independent, the latter involving bypass pathway activation, including dysregulation of downstream signaling cascades. EGFR-independent resistance often renders all EGFR-TKIs ineffective, necessitating further investigation into resistance mechanisms. Case summary We report the case of a 63-year-old Chinese woman diagnosed with synchronous lung adenocarcinoma harboring an EGFR exon 21 far-loop insertion mutation and clear cell renal cell carcinoma (ccRCC). A multidisciplinary team recommended systemic therapy for the lung adenocarcinoma and clinical observation for ccRCC. First-line treatment with bevacizumab plus pemetrexed-carboplatin achieved a progression-free survival (PFS) of 7 months. Second-line treatment with sintilimab and nedaplatin resulted in a PFS of 4.9 months. Third-line therapy with sintilimab and anlotinib proved ineffective. In the fourth line, the patient received furmonertinib, a third-generation EGFR-TKI, based on the FAVOUR trial. This treatment achieved durable disease control with excellent tolerability, yielding a PFS of 27 months and ongoing clinical benefit. Conclusion This case demonstrates that furmonertinib can provide significant clinical benefit to NSCLC patients with complex resistance mechanisms, including those involving the PIK3CA/mTOR pathway. These findings support its potential to overcome EGFR-TKI resistance and warrant further investigation in similar clinical contexts.
Collapse
Affiliation(s)
| | - Peng Wang
- Department Oncology, Yidu Central Hospital of Weifang, Weifang, China
| |
Collapse
|
27
|
Diao X, Guo C, Jin Y, Li B, Gao X, Du X, Chen Z, Jo M, Zeng Y, Ding C, Liu W, Guo J, Li S, Qiu H. Cancer situation in China: an analysis based on the global epidemiological data released in 2024. Cancer Commun (Lond) 2024. [PMID: 39659114 DOI: 10.1002/cac2.12627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 10/26/2024] [Accepted: 10/31/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Cancer remains a major cause of mortality and a significant economic burden in China. Exploring the disparities in cancer patterns and control strategies between China and developed countries may offer valuable insights for policy formulation and enhance cancer management efforts. This study examined the incidence, mortality, and disability-adjusted life year (DALY) burden of cancer in China, and compared these metrics with those observed in the United States (US) and the United Kingdom (UK). METHODS Data on cancer incidence, mortality, and DALYs for China, the US, and the UK were sourced from the GLOBOCAN 2022 online database and the Global Burden of Disease 2021 study (GBD 2021). We utilized Joinpoint regression models to analyze trends in cancer incidence and mortality across these countries, calculating annual percent changes (APCs) and determining the optimal joinpoints. RESULTS In 2022, China recorded around 4,824,703 new cancer cases and 2,574,176 cancer-related deaths, contributing to 71,037,170 DALYs. China exhibited a lower cancer incidence rate compared to the US and the UK. Although cancer-related mortality in China is slightly lower than that in the UK, it is significantly higher than that in the US. Additionally, China experienced significantly higher DALY rates compared to both the US and UK. The cancer landscape in China was also undergoing significant changes, with a rapid rise in the incidence and burden of lung, colorectal, breast, cervical, and prostate cancers. Meanwhile, the incidence and burden of stomach cancer continued to decline. Although the incidence of liver and esophageal cancers was decreasing, the burden of liver cancer was increasing, while the burden of esophageal cancer remained largely unchanged. CONCLUSIONS The cancer profile of China is shifting from that of a developing country to one more typical of a developed country. The ongoing population aging and the rise in unhealthy lifestyles are expected to further escalate the cancer burden in China. Consequently, it is crucial for Chinese authorities to revise the national cancer control program, drawing on successful strategies from developed countries, while also accounting for the regional diversity in cancer types across China.
Collapse
Affiliation(s)
- Xiayao Diao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Chao Guo
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Yukai Jin
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
| | - Bowen Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Xuehan Gao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Xin Du
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Zhenchong Chen
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
| | - Minju Jo
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
| | - Yi Zeng
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
| | - Chao Ding
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
| | - Wenwu Liu
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
| | - Jianrong Guo
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
| | - Shanqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Haibo Qiu
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
28
|
Zhang Q, Pan G, Zhang L, Xu Y, Hao J. The Predictive Value of Monocarboxylate Transporter 4 (MCT4) on Lung Adenocarcinoma Patients Treated with PD-1 Inhibitors. J Inflamm Res 2024; 17:10515-10531. [PMID: 39659754 PMCID: PMC11630727 DOI: 10.2147/jir.s493632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024] Open
Abstract
Purpose Monocarboxylate transporter 4 (MCT4) can influence the amount of lactate in the tumor microenvironment and further control cancer cell proliferation, migration, and angiogenesis. This study aimed to evaluate the predictive value of MCT4 for prognosis and immunotherapy efficacy in advanced lung adenocarcinoma (LUAD). Patients and methods First, bioinformatics analysis was used to assess the relevance of MCT4 for survival and immunotherapy outcomes in LUAD. Subsequently, we performed a retrospective study involving 126 patients with stage IIIb to IV LUAD treated with programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) inhibitors. MCT4 expression in LUAD tissues was detected by immunohistochemistry (IHC), then the patients were divided into high and low expression groups. The differences in the medical records of the two groups were compared using the X2 test. Kaplan-Meier (K-M) method was used for survival analysis. Univariate and multivariate analysis were used to pinpoint independent predictors, and a nomogram was developed based on the significant factors for overall survival (OS) in the multivariate analysis. The predictive ability of the nomogram was evaluated through C-index, receiver operating characteristic (ROC) curve, calibration curve, and decision curve analysis (DCA). Results Both bioinformatics analysis and clinical study revealed that low MCT4 expression was associated with better prognosis and immunotherapy efficacy. Multivariate analysis of clinical characteristics showed that age >65 years, stage IV, high MCT4 expression, neutrophil-to-lymphocyte ratio (NLR)>3, lactate dehydrogenase (LDH)>250 (U/L) and carcinoembryonic antigen (CEA)>5 (ng/mL) were significantly associated with poor prognosis on immunotherapy. These factors were subsequently incorporated into the nomogram model. The C-index value of the model stood at 0.735 (95% CI= 0.662 ~ 0.807), indicating robust predictive performance of the model. The DCA curve showed that the model had a notable clinical application value. Conclusion High expression of MCT4 is associated with poor prognosis and reduced efficacy of immunotherapy in patients with advanced LUAD.
Collapse
Affiliation(s)
- Qinghua Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| | - Guizhen Pan
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| | - Lu Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| | - Yidan Xu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| | - Jiqing Hao
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| |
Collapse
|
29
|
Wang L, Wang R, Wei Z, Wang Y, Chen H, Dong B, Hu X, Ma H, Wang Z, Feng W, Li P, Lin X, Xu Y. Long-term survival and failure patterns in inoperable early-stage non-small cell lung cancer following stereotactic body radiotherapy: a single-institution retrospective study. Sci Rep 2024; 14:30076. [PMID: 39627240 PMCID: PMC11614887 DOI: 10.1038/s41598-024-73177-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 09/16/2024] [Indexed: 12/06/2024] Open
Abstract
This study is to analyse the failure patterns and long-term survival after stereotactic body radiotherapy (SBRT) in patients with T1-3N0M0 inoperable non-small cell lung cancer (NSCLC). Early-stage NSCLC patitents who received SBRT at Zhejiang Cancer Hospital from January 2012 to September 2018 were retrospectively analyzed. The primary endpoint were the patterns of disease progression, which were divided into local recurrence, regional failure, and distant metastasis. Kaplan-Meier method survival analysis was used to calculate overall survival (OS), progression-free survival (PFS). Cox model was used for univariate analysis and multivariate analysis. A total of 215 patients with 224 lesions were enrolled. After the median follow-up time of 50.8 months (1.0-117.9 months), 76 (35.3%) patients progressed, with regional progression occurring in 4 cases (1.8%), local and local-regional progression in 17 cases (7.9%), various distant metastases developing in 55 cases (25.6%). The OS rates at 1, 3, and 5 years were 97.1%, 80.9%, and 63.8%, respectively, with a median OS of 92.2 months (95%CI, 61.5-122.9 months). The PFS rates at 1, 3, and 5 years were 87.5%, 65.9%, and 50.8%, respectively, with a median PFS of 62.2 months (95% CI, 45.0-59.4 months). There was no significant difference in OS (P = 0.832) and PFS (P = 0.672) between the two groups with or without pathology. Multivariate analysis showed that BED and patient age were independent prognostic factors affecting early-stage lung cancer survival (all P < 0.05). Distant metastasis was the main failure pattern of inoperable early-stage NSCLC after SBRT, and the high-risk population should be selected for further systemic treatment.
Collapse
Affiliation(s)
- Lin Wang
- Department of Ultrasonography, Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| | - Ruiqi Wang
- Department of Thoracic Radiotherapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Zhuojun Wei
- Department of Thoracic Radiotherapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yu Wang
- Department of Thoracic Radiotherapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Huan Chen
- Department of Thoracic Radiotherapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Baiqiang Dong
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Xiao Hu
- Department of Thoracic Radiotherapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Honglian Ma
- Department of Thoracic Radiotherapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Zhun Wang
- Department of Thoracic Radiotherapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Wei Feng
- Department of Thoracic Radiotherapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Pu Li
- Department of Radiation Physics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Xiao Lin
- Department of Thoracic Radiotherapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Yujin Xu
- Department of Thoracic Radiotherapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
30
|
Du M, Liu C, Chen L, Li Z, Zhang S, Meng R. Concomitant ALK Fusion and TP53/EGFR Mutation Lead to Adverse Prognostic Outcome. THE CLINICAL RESPIRATORY JOURNAL 2024; 18:e70041. [PMID: 39681089 DOI: 10.1111/crj.70041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 11/11/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024]
Abstract
Lung cancer treatment has evolved at the molecular level. Detecting the presence of driver genes in lung cancer fundamentally alters the choice of therapeutic regimens and the outcome of this disease. ALK fusion mutation is one of the most important mutations in nonsmall cell lung cancer (NSCLC). Also, it often has other coexisting mutation types. TP53 is the most common coexisting mutation type, whereas the EGFR/ALK coexisting mutation type is extremely rare. There is still no definite conclusion about the impact of the multimutation and best treatment options for NSCLC patients with advanced multimutation. In this study, we report three cases of NSCLC with ALK fusion mutations as well as ALK combined with TP53 mutations and ALK combined with EGFR mutations. Combining cases from our oncology center and previous literature, we found that NSCLC patients with coexisting ALK fusion mutations and other mutations have poorer response to targeted therapy and poorer prognosis, and we also compared the efficacy rates of various types of coexisting mutations for different treatment regimens. Therefore, this review can help to evaluate the prognosis of NSCLC patients with coexisting mutations and the efficacy of targeted therapies and to find more favorable treatment options for patients with this type of coexisting mutations.
Collapse
Affiliation(s)
- Mingyuan Du
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cuiwei Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Leichong Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenyu Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sijia Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Meng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
Fang F, Zhao M, Jin X, Dong Z, Wang J, Meng J, Xie S, Shi W. RETRACTED ARTICLE: Upregulation of MCL-1 by LUCAT1 through interacting with SRSF1 promotes the migration and invasion in non-small cell lung carcinoma. Mol Cell Biochem 2024; 479:3305. [PMID: 37747676 DOI: 10.1007/s11010-023-04851-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 09/04/2023] [Indexed: 09/26/2023]
Affiliation(s)
- Fang Fang
- The Second Ward of Oncology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, No. 327, Xianhu Avenue, Qingxiu District, Nanning, 530001, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Mei Zhao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530001, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xiaowei Jin
- Department of Traditional Chinese Medicine, Yunnan Cancer Hospital, Kunming, 650018, Yunnan Province, People's Republic of China
| | - Zhixin Dong
- The Second Ward of Oncology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, No. 327, Xianhu Avenue, Qingxiu District, Nanning, 530001, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Jiaxiao Wang
- The Second Ward of Oncology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, No. 327, Xianhu Avenue, Qingxiu District, Nanning, 530001, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Jinming Meng
- Guangxi University of Chinese Medicine, Nanning, 530001, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Sheng Xie
- Preventive Treatment of Disease Center, The First Affiliated Hospital of Guangxi University of Chinese Medicine, No. 89-9, Dongge Road, Qingxiu District, Nanning, 530023, Guangxi Zhuang Autonomous Region, People's Republic of China.
| | - Wei Shi
- The Second Ward of Oncology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, No. 327, Xianhu Avenue, Qingxiu District, Nanning, 530001, Guangxi Zhuang Autonomous Region, People's Republic of China.
| |
Collapse
|
32
|
Zhang N, Bai T, Jiang Y, Zhu K, Yao L, Ji J, Huang Q. Role of SFRP5 in Non-Small Cell Lung Cancer and Its Correlation with SUV of 18F-FDG PET-CT. J INVEST SURG 2024; 37:2381722. [PMID: 39074839 DOI: 10.1080/08941939.2024.2381722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/06/2024] [Accepted: 07/13/2024] [Indexed: 07/31/2024]
Abstract
Aim: This study aimed to evaluate the relationship between secreted frizzled-related protein 5 (SFRP5) expression and fluorine 18-fluoro-deoxyglucose (18 F-FDG) uptake imaged with positron emission tomography/tomography (PET/CT) in patients with non-small cell lung cancer (NSCLC). In addition, we sought to elucidate the potential role and mechanism of action of SFRP5 in NSCLC.Materials and methods: The maximum standardized uptake value (SUVmax) of the lesions was calculated. SFRP5 expression was analyzed using quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). The correlation between SFRP5 expression and SUVmax was evaluated using Pearson's correlation analysis. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), flow cytometry, wound healing, and transwell assays were used to analyze cell viability, apoptosis, migration, and invasion, respectively.Results and conclusion: The results indicated that the SUVmax was higher in patients with NSCLC than that in healthy volunteers. Moreover, SFRP5 expression was lower in tissues from the four types of NSCLC than that in the adjacent normal tissues. SUVmax negatively correlated with SFRP5 expression in the four types of NSCLC. In addition, up-regulation of SFRP5 decreased the viability, migration, and invasion abilities, and increased apoptosis of NSCLC cells. Furthermore, SFRP5 inhibited the Wnt/β-catenin pathway in NSCLC cells. In conclusion, SFRP5 modulates the biological behaviors of NSCLC through Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Na Zhang
- Radiographic Imaging Center, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Tian Bai
- Radiographic Imaging Center, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Yunfei Jiang
- Respiratory Department, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Kun Zhu
- Cardiothoracic Surgery Department, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Lan Yao
- Nuclear Medicine Department, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Jia Ji
- Stomatology Department, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Qicheng Huang
- Radiographic Imaging Center, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
33
|
Chen S, Yang H, Hu Z, Jin J, Xiong X, Zhang Z, Xie C. Deacetylation by SIRT6 increases the stability of GILZ to suppress NSCLC cell migration and invasion. Cell Signal 2024; 124:111414. [PMID: 39288887 DOI: 10.1016/j.cellsig.2024.111414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
Glucocorticoid-induced leucine zipper (GILZ) plays a role in cancer cell proliferation in several tumor types. However, in our present study, GILZ was demonstrated to be a metastasis regulator but not a proliferation regulator in non-small cell lung cancer (NSCLC). The overexpression of GILZ had no significant effect on the proliferation of NSCLC cells but inhibited their metastasis by targeting the epithelial-mesenchymal transition pathway. The deacetylase SIRT6, a key regulator of protein stability, can enhance the stability of the GILZ protein by mediating its deacetylation, which prevents ubiquitination and degradation. This process ultimately enhances the inhibitory effect of GILZ on the migration and invasion of NSCLC cells. Thus, GILZ may be a promising new therapeutic target for tumor metastasis.
Collapse
Affiliation(s)
- Shiyin Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, PR China; Department of Pathology, Jiangxi Maternal & Child Health Hospital, Nanchang, Jiangxi, PR China
| | - Huaping Yang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, PR China
| | - Zhenghui Hu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, PR China
| | - Jiangbo Jin
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, PR China
| | - Xiangyang Xiong
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, PR China
| | - Ziyu Zhang
- Department of Pathology, Jiangxi Maternal & Child Health Hospital, Nanchang, Jiangxi, PR China
| | - Caifeng Xie
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, PR China.
| |
Collapse
|
34
|
Lee SH, Menis J, Kim TM, Kim HR, Zhou C, Kurniawati SA, Prabhash K, Hayashi H, Lee DDW, Imasa MS, Teh YL, Yang JCH, Reungwetwattana T, Sriuranpong V, Wu CE, Ang Y, Sabando M, Thiagarajan M, Mizugaki H, Noronha V, Yulianti M, Zhang L, Smyth E, Yoshino T, Park JO, Pentheroudakis G, Park S, Peters S, Ahn JB, Popat S. Pan-Asian adapted ESMO Clinical Practice Guidelines for the diagnosis, treatment and follow-up of patients with oncogene-addicted metastatic non-small-cell lung cancer. ESMO Open 2024; 9:103996. [PMID: 39615406 DOI: 10.1016/j.esmoop.2024.103996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/14/2024] [Accepted: 10/28/2024] [Indexed: 12/24/2024] Open
Abstract
The European Society for Medical Oncology (ESMO) Clinical Practice Guidelines for the diagnosis, treatment and follow-up of patients with oncogene-addicted metastatic non-small-cell lung cancer (mNSCLC), published in January 2023, was modified according to previously established standard methodology, to produce the Pan-Asian adapted (PAGA) ESMO consensus guidelines for the management of Asian patients with oncogene-addicted mNSCLC. The adapted guidelines presented in this manuscript represent the consensus opinions reached by a panel of Asian experts in the treatment of patients with oncogene-addicted mNSCLC representing the oncological societies of China (CSCO), Indonesia (ISHMO), India (ISMPO), Japan (JSMO), Korea (KSMO), Malaysia (MOS), the Philippines (PSMO), Singapore (SSO), Taiwan (TOS) and Thailand (TSCO), co-ordinated by ESMO and the Korean Society for Medical Oncology (KSMO). The voting was based on scientific evidence and was independent of the current treatment practices, drug access restrictions and reimbursement decisions in the different regions of Asia. The latter are discussed separately in the manuscript. The aim is to provide guidance for the optimisation and harmonisation of the management of patients with oncogene-addicted mNSCLC across the different regions of Asia, drawing on the evidence provided by both Western and Asian trials, while respecting the differences in screening practices, molecular profiling and age and stage at presentation. Attention is drawn to the disparity in the drug approvals and reimbursement strategies between the different regions of Asia.
Collapse
Affiliation(s)
- S-H Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| | - J Menis
- Medical Oncology Department, University and Hospital Trust of Verona, Verona, Italy
| | - T M Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - H R Kim
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - C Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - S A Kurniawati
- Department of Internal Medicine, Division of Hematology and Medical Oncology, Dharmais Cancer Hospital, National Cancer Center, Jakarta, Indonesia
| | - K Prabhash
- Department of Medical Oncology, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, India
| | - H Hayashi
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | - D D-W Lee
- Department of Clinical Oncology, University of Malaya, Kuala Lumpur, Malaysia
| | - M S Imasa
- Department of Thoracic Oncology, Lung Center of the Philippines, Quezon City, Philippines
| | - Y L Teh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - J C-H Yang
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - T Reungwetwattana
- Division of Medical Oncology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - V Sriuranpong
- Division of Medical Oncology, Faculty of Medicine, Chulalongkorn University and the King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - C-E Wu
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Y Ang
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - M Sabando
- Internal Medicine Department, Bicol Regional Hospital and Medical Center, Legazpi City, Albay, Philippines
| | - M Thiagarajan
- Department of Radiotherapy and Oncology, Kuala Lumpur Hospital, Kuala Lumpur, Malaysia
| | - H Mizugaki
- Department of Respiratory Medicine, NHO Hokkaido Cancer Center, Sapporo, Japan
| | - V Noronha
- Department of Medical Oncology, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, India
| | - M Yulianti
- Division of Respirology and Critical Care, Department of Internal Medicine, Faculty of Medicine Universitas Indonesia-Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - L Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - E Smyth
- Department of Oncology, Oxford University Hospital NHS Foundation Trust, Oxford, UK
| | - T Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - J O Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | - S Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - S Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - J B Ahn
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - S Popat
- Royal Marsden NHS Trust, London, UK
| |
Collapse
|
35
|
Xu H, Liang Y, Tang W, Yang X, Du X. Efficacy and safety of anlotinib combined with S‑1 as a third‑ or later‑line treatment for advanced non‑small cell lung cancer in China: A systematic review and meta‑analysis. Oncol Lett 2024; 28:613. [PMID: 39493432 PMCID: PMC11528180 DOI: 10.3892/ol.2024.14746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/19/2024] [Indexed: 11/05/2024] Open
Abstract
Anlotinib is presently used as a third-line treatment for non-small cell lung cancer. However, it is not yet reported whether combining anlotinib with S-1 as a third- or later-line treatment offers superior outcomes compared with anlotinib alone. The present meta-analysis aimed to address this question by systematically searching the PubMed, Embase, Web of Science, Cochrane Library, CMB and China National Knowledge Infrastructure databases for eligible studies published from the establishment of the database to January 10, 2024. Primary outcomes of interest included progression-free survival (PFS), overall survival (OS), objective response rate (ORR), disease control rate (DCR) and the incidence of adverse effects, which were presented as hazard ratios and 95% CIs. The present analysis included 5 retrospective studies with a total of 317 patients and compared the outcomes of patients treated with a combination of anlotinib and S-1 (experimental group) compared with anlotinib alone (control group). The combination treatment significantly improved PFS, OS, ORR and DCR in the experimental group compared with the control group. Bone marrow suppression and fatigue were significantly higher in the experimental group compared with the control group. However, incidences of hypertension, proteinuria, gastrointestinal adverse reactions, hepatic and renal insufficiency and functional hand-foot syndrome were higher in the control group compared with the experimental group, but there was no statistical significance. In summary, combining anlotinib with S-1 may be more effective compared with anlotinib alone for treating advanced non-small cell lung cancer. Despite the higher incidence of adverse reactions with the combination therapy, these reactions could be considered manageable and controllable.
Collapse
Affiliation(s)
- Haonan Xu
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
- Department of Oncology, National Health Commission Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, Mianyang, Sichuan 621000, P.R. China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang Central Hospital, Mianyang, Sichuan 621000, P.R. China
| | - Yuwen Liang
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
- Department of Oncology, National Health Commission Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, Mianyang, Sichuan 621000, P.R. China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang Central Hospital, Mianyang, Sichuan 621000, P.R. China
| | - Wenqiang Tang
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
- Department of Oncology, National Health Commission Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, Mianyang, Sichuan 621000, P.R. China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang Central Hospital, Mianyang, Sichuan 621000, P.R. China
| | - Xiongxin Yang
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Xiaobo Du
- Department of Oncology, National Health Commission Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, Mianyang, Sichuan 621000, P.R. China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang Central Hospital, Mianyang, Sichuan 621000, P.R. China
| |
Collapse
|
36
|
Gao J, Liang C, Yin J, Bai Y, Hu D. Discovery of Palbociclib as a potent c-Myc G4 stabilizer for lung cancer treatment using molecular docking, molecular dynamics simulation, and in vitro activity evaluation. Mol Divers 2024; 28:3965-3977. [PMID: 38246949 DOI: 10.1007/s11030-023-10789-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/02/2023] [Indexed: 01/23/2024]
Abstract
Despite significant progress in lung cancer treatment, this disease remains a prevalent and serious global malignancy, leading to high rates of illness and death. Urgent research is needed to discover new or alternative therapies that can improve clinical outcomes for lung cancer patients. In our study, we successfully demonstrated the effectiveness of Palbociclib, a CDK4/6 inhibitor, in suppressing the growth of lung cancer cells. The IC50 values obtained were 11.00 μM and 11.74 μM for H1299 and A549 cells, respectively. Furthermore, our findings indicate that Palbociclib may possess strong c-Myc G4 stabilizing properties by significantly reducing both protein and mRNA expression levels of c-Myc. Additionally, Palbociclib induces apoptosis and causes cell cycle arrest at the G2/M phase in two cells. Through circular dichroism (CD), molecular docking, and molecular dynamics (MD) simulation, we have provided evidence that Palbociclib enhances the structural stability of c-Myc G4 while exhibiting a high binding affinity to its ligand's binding site on c-Myc G4. These results suggest that Palbociclib holds promise as a novel c-Myc G4 stabilizer for treating cancers associated with abnormal c-Myc activity; further optimization and development are warranted.
Collapse
Affiliation(s)
- Jian Gao
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Chao Liang
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Jiacheng Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Ying Bai
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Dong Hu
- School of Medicine, Anhui University of Science and Technology, Huainan, China.
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, 232001, China.
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institute, Huainan, 232001, China.
- Key Laboratory of Industrial Dust Prevention and Control & Occupational Safety and Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, 232001, China.
| |
Collapse
|
37
|
Zhang B, Luo D, Xiang L, Chen J, Fang T. Investigating the anti-cancer potential of sulfatase 1 and its underlying mechanism in non-small cell lung cancer. Cytojournal 2024; 21:52. [PMID: 39737127 PMCID: PMC11683397 DOI: 10.25259/cytojournal_71_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/29/2024] [Indexed: 01/01/2025] Open
Abstract
Objective Patients with non-small cell lung cancer (NSCLC) have poor prognoses. Sulfatase 1 (SULF1) is an extracellular neutral sulfatase and is involved in multiple physiological processes. Hence, this study investigated the function and possible mechanisms of SULF1 in NSCLC. Material and Methods Difference in SULF1 expression level between tumors and normal lung tissues was analyzed through bioinformatics and clinical sampling, and the effects of SULF1 expression on prognosis were investigated through Kaplan-Meier analysis. SULF1 level in NSCLC cells was modulated through small interfering ribonucleic acid interference. NSC228155, which is an epidermal growth factor receptor (EGFR)/mitogen-activated protein kinase (MAPK) signaling pathway agonist, was for handling NSCLC cells. SULF1 expression level was tested through quantitative reverse transcriptase real-time polymerase chain reaction. Cell proliferation, migration, and invasion were evaluated with cell counting kit-8, 5-ethynyl-2-deoxyuridine, and transwell assays, and the levels of epithelial-to-mesenchymal transition (EMT)- and EGFR/MAPK pathway-related proteins were detected through Western blot. Results Bioinformatics and clinical samples showed that NSCLC tumor tissues had elevated SULF1 expression levels relative to those of normal tissues (P < 0.05). Patients with NSCLC and high SULF1 expression levels experienced poorer prognosis than those of low SULF1 expression levels (P < 0.05). SULF1 knockdown repressed the malignant biological behavior, including proliferation, migration, and invasion, of the NSCLC cells (P < 0.05). Mechanistically, SULF1 knockdown augmented E-cadherin level and abated N-cadherin and vimentin protein levels (P < 0.05). These results confirmed that EMT was inhibited. In addition, the knockdown of SULF1 reduced the phosphorylation of EGFR, extracellular signal-regulated kinase, p38 MAPK and c-Jun N-terminal kinase, and NSC228155 partially reversed these changes, which were affected by SULF1 knockdown. Meanwhile, NSC228155 partially reversed the inhibition of EMT, migration, and invasion affected by SULF1 knockdown. Conclusion SULF1 knockdown inhibits the proliferation, migration, invasion, and EMT of NSCLC cells by inactivating EGFR/MAPK pathway.
Collapse
Affiliation(s)
- Bingling Zhang
- Department of Disease Control and Prevention, Zhangqiao Branch, Ningbo Ninth Hospital Medical Health Group, Ningbo, China
| | - Daping Luo
- Department of Prevention and Healthcare, Hongtang Branch, Ningbo Ninth Hospital Medical Health Group, Ningbo, China
| | - Lan Xiang
- Department of Doctor-Patient Communication, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Jun Chen
- Department of Disease Control and Prevention, Zhangqiao Branch, Ningbo Ninth Hospital Medical Health Group, Ningbo, China
| | - Ting Fang
- Medical College, Ningbo University Health Science Center, Ningbo, China
| |
Collapse
|
38
|
Liu M, Tang B, Xiang R, Hu P, Xu C, Hu L, Li Q. Aberrant expression of MRAS and HEG1 as the biomarkers for osimertinib resistance in LUAD. Discov Oncol 2024; 15:678. [PMID: 39560891 DOI: 10.1007/s12672-024-01552-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
Epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) are the most applied targeted therapy for EGFR-mutant lung adenocarcinoma (LUAD). The third-generation EGFR-TKI, osimertinib, is widely used throughout lung cancer treatment, with single or combination modes. One of the main barriers in osimertinib treatment is the acquired resistance and mechanisms are not fully understood. Gene expression other than genetic mutations might predict drug response and mediate resistance occurrence. We analyzed six datasets of osimertinib-resistant LUAD cells from the Gene Expression Omnibus (GEO) database and identified two hub genes, named MRAS and HEG1. We found that the expression mode of MRAS/HEG1 in LUAD was osimertinib-dependent and contributed to drug resistance. We also explored potential mechanisms of hub genes related osimertinib resistance and emphasized the M2 infiltration involved. Moreover, potential therapeutic agents conquering MRAS/HEG1-related resistance were also identified. In conclusion, MRAS and HEG1 might be responsible for osimertinib resistance and could be promising prognostic biomarkers for osimertinib response in LUAD, which might provide insights into therapeutic strategies.
Collapse
Affiliation(s)
- Mingxin Liu
- Department of Thoracic Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610042, China
| | - Bo Tang
- Department of Oncology and Cancer Institute, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Run Xiang
- Department of Thoracic Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610042, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan University, Chengdu, 610042, China
| | - Peihong Hu
- Department of Thoracic Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610042, China
| | - Chuan Xu
- Department of Oncology and Cancer Institute, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Yu-Yue Pathology Scientific Research Center, Chongqing, 400039, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Lanlin Hu
- Department of Oncology and Cancer Institute, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Yu-Yue Pathology Scientific Research Center, Chongqing, 400039, China.
- Jinfeng Laboratory, Chongqing, 401329, China.
| | - Qiang Li
- Department of Thoracic Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610042, China.
| |
Collapse
|
39
|
Li N, Zhu X, Zhang H, Yang X, Shao M, Cui S, Lin C. Exploring the Target Genes of Fucosylated Chondroitin Sulfate in Treating Lung Adenocarcinoma Based on the Integration of Bioinformatics Analysis, Molecular Docking, and Experimental Verification. ACS OMEGA 2024; 9:46312-46322. [PMID: 39583738 PMCID: PMC11579779 DOI: 10.1021/acsomega.4c07295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/25/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024]
Abstract
Fucosylated chondroitin sulfate (FCS), extracted from sea cucumbers' body walls, has been found to inhibit the proliferation of lung adenocarcinoma (LUAD) cells. However, there have been few studies of the associated drug targets. This study combined bioinformatics analysis and molecular docking to screen the main targets of FCS intervention in LUAD. Moreover, an experimental validation was performed. First, we downloaded the LUAD gene data set from The Cancer Genome Atlas (TCGA) database and the cisplatin (DDP) resistance gene data set of LUAD A549 cells from the Gene Expression Omnibus (GEO) database. Nine significant genes (PLK1, BUB1, CDK1, CDC20, CCNB1, BUB1B, KIF11, CCNB2, and DLAGP5) were identified by bioinformatics analysis, and these nine genes overlapped in both data sets. Then, molecular docking results showed that FCS had a better affinity with target proteins BUB1 and PLK1. Further experimental verification revealed that FCS inhibited the growth of A549 cells and increased the sensitivity of A549 cells to DDP. Quantitative real-time polymerase chain reaction (qRT-PCR) revealed that A549 cells treated with FCS exhibited down-regulated BUB1 and PLK1 mRNA expression. At the same time, FCS+DDP treatment resulted in a more significant reduction in BUB1 and PLK1 mRNA expression than DDP or FCS treatment alone. These findings reveal potential targets of FCS for LUAD and provide clues for the development of FCS as a potential anticancer agent.
Collapse
Affiliation(s)
- Nana Li
- Department
of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Xinhong Zhu
- Department
of International Medicine, Qingdao Municipal
Hospital Group, Qingdao 266071, China
| | - Hua Zhang
- Department
of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Xiaohui Yang
- Department
of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Mingju Shao
- Department
of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Shichao Cui
- Department
of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Cunzhi Lin
- Department
of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| |
Collapse
|
40
|
Guo H, Wang H, Gao M, Deng H, Zhang Y, Gong J, Zhang W. Harnessing the CD44-targeted delivery of self-assembled hyaluronan nanogel to reverse the antagonism between Cisplatin and Gefitinib in NSCLC cancer therapy. Carbohydr Polym 2024; 344:122521. [PMID: 39218565 DOI: 10.1016/j.carbpol.2024.122521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/10/2024] [Accepted: 07/17/2024] [Indexed: 09/04/2024]
Abstract
The combination of the standard platinum-based chemotherapy with EGFR-tyrosine kinase inhibitor Gefitinib (Gef) principally boosts the anticancer efficacy of advanced non-small cell lung cancer (NSCLC) through non-overlapping mechanisms of action, however the clinical trials of cisplatin (Cis) and Gef combination failed to show a therapeutic improvement likely due to compromised cellular influx of Cis with the Gef interference. To overcome the antagonism between Cis and Gef in anti-NSCLC therapy, here we demonstrated a self-targeted hyaluronan (HA) nanogel to facilitate the anticancer co-delivery by utilizing the HA's intrinsic targeting towards CD44, a receptor frequently overexpressed on lung cancer cells. The co-assembly between HA, Cis and Gef generated a HA/Cis/Gef nanogel of 177.8 nm, featuring a prolonged drug release. Unlike the Gef inhibited the Cis uptake, the HA/Cis/Gef nanogel efficiently facilitated the drug internalization through CD44-targeted delivery as verified by HA competition and CD44 knocking down in H1975 NSCLC model both in vitro and in vivo. Moreover, the HA/Cis/Gef nanogel significantly improved the anticancer efficacy and meanwhile diminished the side effects in reference to the combination of free Cis and Gef. This CD44-targeted HA/Cis/Gef nanogel provided a potent strategy to advance the platinum-based combination therapy towards optimized NSCLC therapy.
Collapse
Affiliation(s)
- Hua Guo
- State Key Laboratory of Molecular Oncology and Department of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China
| | - Huimin Wang
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Menghan Gao
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Hong Deng
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Yiyi Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Jianan Gong
- The Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Weiqi Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China.
| |
Collapse
|
41
|
Wang J, Xu Y, Wang T. Efficacy Analysis of Bronchial Arterial Chemoembolization for Nonsmall Cell Lung Cancer: A Systematic Review and Meta-Analysis. Cancer Biother Radiopharm 2024. [PMID: 39527099 DOI: 10.1089/cbr.2024.0141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Objective: This study aims to comprehensively evaluate the efficacy and safety of bronchial arterial chemoembolization (BACE) in the treatment of advanced nonsmall cell lung cancer (NSCLC) through a meta-analysis of single-group rate, providing evidence-based guidance for clinical treatment. Materials and Methods: A systematic search was conducted in PubMed, the Cochrane Library, Embase, and Web of Science databases for relevant studies up to January 15, 2024. Inclusion criteria encompassed single-arm or multi-arm studies of nonrandomized controlled trials, observational studies, and single-arm studies in English language, focusing on NSCLC patients treated with BACE. Data extraction, quality assessment, and statistical analysis were performed following predefined protocols. Results: In total, 172 articles were initially retrieved, with 11 studies meeting the inclusion criteria. The included studies comprised 510 patients. Meta-analysis revealed significant heterogeneity among studies for median progression-free survival (PFS), median overall survival (OS), objective response rate, and disease control rate. The combined median PFS was 6.87 months (95% confidence interval [CI] 5.30-8.44), and the combined median OS was 13.68 months (95% CI 10.69-16.67). Subgroup analysis based on intervention measures demonstrated varying efficacy outcomes. Adverse reactions associated with BACE were generally mild, with no reports of grade 3 or higher adverse events. Conclusion: BACE emerges as a promising treatment modality for advanced NSCLC, exhibiting favorable efficacy and safety profiles.
Collapse
Affiliation(s)
- Jiayao Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- The Center for Biomedical Research, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yahan Xu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- The Center for Biomedical Research, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Tao Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
42
|
Ge X, Shen Z, Yin Y. Comprehensive review of LncRNA-mediated therapeutic resistance in non-small cell lung cancer. Cancer Cell Int 2024; 24:369. [PMID: 39522033 PMCID: PMC11549762 DOI: 10.1186/s12935-024-03549-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Long non-coding RNAs (lncRNAs) are emerging as crucial regulators of gene expression through diverse mechanisms, including regulation of protein localization, sequestration of miRNAs, recruitment of chromatin modifiers, and modulation of signaling pathways. Accumulating evidence highlights their pivotal roles in tumor initiation, progression, and the development of therapeutic resistance. In this review, we comprehensively summarized the existing literature to identify lncRNAs associated with treatment responses in non-small cell lung cancer (NSCLC). Specifically, we categorized these lncRNAs based on their mechanisms of action in mediating resistance to chemotherapy, targeted therapy, and radiotherapy. Our analysis revealed that aberrant expression of various lncRNAs contributes to the development, metastasis, and therapeutic resistance in NSCLC, ultimately leading to poor clinical outcomes. By elucidating the intricate mechanisms through which lncRNAs modulate therapeutic responses, this review aims to provide mechanistic insights into the heterogeneous treatment outcomes observed in NSCLC patients and unveil potential therapeutic targets for overcoming drug resistance.
Collapse
Affiliation(s)
- Xin Ge
- Peking University First Hospital, Beijing, 100034, China
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking-Tsinghua Center of Life Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Zichu Shen
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking-Tsinghua Center of Life Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yuxin Yin
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking-Tsinghua Center of Life Sciences, Peking University Health Science Center, Beijing, 100191, China.
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| |
Collapse
|
43
|
Wu S, Dong C, Hu C, Hui K, Jiang X. Case report: Combination therapy of envafolimab with endostar for advanced non-small cell lung cancer with low PD-L1 expression. Front Oncol 2024; 14:1437260. [PMID: 39575420 PMCID: PMC11578815 DOI: 10.3389/fonc.2024.1437260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 10/18/2024] [Indexed: 11/24/2024] Open
Abstract
In the management of advanced non-squamous non-small cell lung cancer (NSCLC) without driver gene mutations, the current therapeutic strategies encompass chemotherapy, chemotherapy combined with anti-angiogenic therapy, and chemotherapy combined with immunotherapy. For patients with high programmed death-ligand 1(PD-L1) expression, monotherapy with immune checkpoint inhibitors is a viable option. Recognizing that some patients cannot tolerate or decline chemotherapy, clinical practice has introduced non-chemotherapeutic treatment regimens, which have shown promising results. This article presents a clinical case of advanced NSCLC with low PD-L1 expression and negative driver gene mutations. The patient was treated with a chemotherapy-free regimen combining envafolimab with endostar. After 17 months of follow-up, both the primary tumor and metastatic lesions exhibited significant reduction, and no notable adverse reactions were observed. This case demonstrates the efficacy of envafolimab combined with endostar in the treatment of advanced NSCLC. This regimen enhances treatment safety and patient compliance, potentially offering a novel therapeutic option for patients with advanced NSCLC characterized by low PD-L1 expression and negative driver gene mutations.
Collapse
Affiliation(s)
- Shuo Wu
- Department of Radiation Oncology, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, China
| | - Changhong Dong
- Department of Radiation Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, China
| | - Chenxi Hu
- Department of Radiation Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, China
| | - Kaiyuan Hui
- Department of Radiation Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, China
| | - Xiaodong Jiang
- Department of Radiation Oncology, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, China
- Department of Radiation Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, China
| |
Collapse
|
44
|
Li H, Dai X, Zhou J, Wang Y, Zhang S, Guo J, Shen L, Yan H, Jiang H. Mitochondrial dynamics in pulmonary disease: Implications for the potential therapeutics. J Cell Physiol 2024; 239:e31370. [PMID: 38988059 DOI: 10.1002/jcp.31370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
Mitochondria are dynamic organelles that continuously undergo fusion/fission to maintain normal cell physiological activities and energy metabolism. When mitochondrial dynamics is unbalanced, mitochondrial homeostasis is broken, thus damaging mitochondrial function. Accumulating evidence demonstrates that impairment in mitochondrial dynamics leads to lung tissue injury and pulmonary disease progression in a variety of disease models, including inflammatory responses, apoptosis, and barrier breakdown, and that the role of mitochondrial dynamics varies among pulmonary diseases. These findings suggest that modulation of mitochondrial dynamics may be considered as a valid therapeutic strategy in pulmonary diseases. In this review, we discuss the current evidence on the role of mitochondrial dynamics in pulmonary diseases, with a particular focus on its underlying mechanisms in the development of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), chronic obstructive pulmonary disease (COPD), asthma, pulmonary fibrosis (PF), pulmonary arterial hypertension (PAH), lung cancer and bronchopulmonary dysplasia (BPD), and outline effective drugs targeting mitochondrial dynamics-related proteins, highlighting the great potential of targeting mitochondrial dynamics in the treatment of pulmonary disease.
Collapse
Affiliation(s)
- Hui Li
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xinyan Dai
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Junfu Zhou
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Yujuan Wang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Shiying Zhang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Jiacheng Guo
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Lidu Shen
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Hengxiu Yan
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Huiling Jiang
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| |
Collapse
|
45
|
Song Z, Zhang W, Zhu M, Wang Y, Zhou D, Cao X, Geng X, Zhou S, Li Z, Wei K, Chen L. Unveiling Varied Cell Death Patterns in Lung Adenocarcinoma Prognosis and Immunotherapy Based on Single-Cell Analysis and Machine Learning. J Cell Mol Med 2024; 28:e70218. [PMID: 39602465 PMCID: PMC11601877 DOI: 10.1111/jcmm.70218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/12/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Programmed cell death (PCD) pathways hold significant influence in the etiology and progression of a variety of cancer forms, particularly offering promising prognostic markers and clues to drug sensitivity for lung adenocarcinoma (LUAD) patients. We employed single-cell analysis to delve into the functional role of PCD within the tumour microenvironment (TME) of LUAD. Employing a machine learning framework, a PCD-related signature (PCDS) was constructed utilising a comprehensive data set. The PCDS exhibited superior prognostic performance compared with the 140 previously established prognostic models for LUAD. Subsequently, patients were stratified into high-risk and low-risk groups based on their risk scores derived from the PCDS, with the high-risk group exhibiting significantly lower overall survival (OS) rates than the low-risk group. Furthermore, the risk subgroups were compared for differences in pathway enrichment, genomic alterations, tumour immune microenvironment (TIME), immunotherapy and drug sensitivity. The low-risk group displayed a more inflamed TIME, potentially leading to a more favourable response to immunotherapy. For the high-risk group, potential effective small molecule drugs were identified, and the drug sensitivity were evaluated. Immunohistochemistry and quantitative real-time polymerase chain reaction assays (qRT-PCR) confirmed notable upregulation of the expression levels of PCD-associated genes MKI67, TYMS and LYPD3 in LUAD tissues. In vitro experimental findings demonstrated a marked decrease in the proliferative and migratory capacities of LUAD cells upon knockdown of MKI67. Conclusively, we successfully constructed the PCDS, providing important assistance for prognosis prediction and treatment optimisation of LUAD patients.
Collapse
Affiliation(s)
- Zipei Song
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Weiran Zhang
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Miaolin Zhu
- Department of OncologyThe Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer ResearchNanjingChina
| | - Yuheng Wang
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Dingye Zhou
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Xincen Cao
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Xin Geng
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Shengzhe Zhou
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Zhihua Li
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Ke Wei
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Liang Chen
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
46
|
Wu A, Yang H, Xiao T, Gu W, Li H, Chen P. COPZ1 regulates ferroptosis through NCOA4-mediated ferritinophagy in lung adenocarcinoma. Biochim Biophys Acta Gen Subj 2024; 1868:130706. [PMID: 39181476 DOI: 10.1016/j.bbagen.2024.130706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/05/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Ferroptosis, a type of autophagy-dependent cell death, has been implicated in the pathogenesis of lung adenocarcinoma (LUAD). This study aimed to investigate the involvement of coatomer protein complex I subunit zeta 1 (COPZ1) in ferroptosis and ferritinophagy in LUAD. METHODS Publicly available human LUAD sample data were obtained from the TCGA database to analyze the association of COPZ1 expression with LUAD grade and patient survival. Clinical samples of LUAD and para-carcinoma tissues were collected. COPZ1-deficient LUAD cell model and xenograft model were established. These models were analyzed to evaluate tumor growth, lipid peroxidation levels, mitochondrial structure, autophagy activation, and iron metabolism. RESULTS High expression of COPZ1 was indicative of malignancy and poor overall survival. Clinical LUAD tissues showed increased COPZ1 expression and decreased nuclear receptor coactivator 4 (NCOA4) expression. COPZ1 knockdown inhibited xenograft tumor growth and induced apoptosis. COPZ1 knockdown elevated the levels of ROS, Fe2+ and lipid peroxidation. COPZ1 knockdown also caused mitochondrial shrinkage. Liproxstatin-1, deferoxamine, and z-VAD-FMK reversed the effects of COPZ1 knockdown on LUAD cell proliferation and ferroptosis. Furthermore, COPZ1 was directly bound to NCOA4. COPZ1 knockdown restricted FTH1 expression and promoted NCOA4 and LC3 expression. NCOA4 knockdown reversed the regulation of iron metabolism, lipid peroxidation, and mitochondrial structure induced by COPZ1 knockdown. COPZ1 knockdown induced the translocation of ferritin to lysosomes for degradation, whereas NCOA4 knockdown disrupted this process. CONCLUSION This study provides novel evidence that COPZ1 regulates NCOA4-mediated ferritinophagy and ferroptosis. These findings provide new insights into the pathogenesis and potential treatment of LUAD.
Collapse
Affiliation(s)
- Anbang Wu
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Hongmin Yang
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Tengfei Xiao
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wangnin Gu
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - He Li
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; College of pharmacy, Changsha Medical University, Changsha 410219, China.
| | - Pan Chen
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.
| |
Collapse
|
47
|
Liu X, Wang J, Yang Z, Xie Q, Diao X, Yao X, Huang S, Chen R, Zhao Y, Li T, Jiang M, Lou Z, Huang C. Upregulated DNMT3a coupling with inhibiting p62-dependent autophagy contributes to NNK tumorigenicity in human bronchial epithelial cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 286:117157. [PMID: 39393198 DOI: 10.1016/j.ecoenv.2024.117157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/27/2024] [Accepted: 10/04/2024] [Indexed: 10/13/2024]
Abstract
NNK, formally known as 4-(methyl nitrosamine)-1-(3-pyridyl)-1-butanoe, is a potent chemical carcinogen prevalent in cigarette smoke and is a key contributor to the development of human lung adenocarcinomas. On the other hand, autophagy plays a complex role in cancer development, acting as a "double-edged sword" whose impact varies depending on the cancer type and stage. Despite this, the relationship between autophagy and NNK-induced lung carcinogenesis remains largely unexplored. Our current study uncovers a marked reduction in p62 protein expression in both lung adenocarcinomas and lung tissues of mice exposed to cigarette smoke. Interestingly, this reduction appears to be contingent upon the activity of extrahepatic cytochrome P450 (CYP450), revealing that NNK metabolic activation by CYP450 enzyme escalates its potential to induce p62 downregulation. Further mechanistic investigations reveal that NNK suppresses autophagy by accelerating the degradation of p62 mRNA, thereby promoting the malignant transformation of human bronchial epithelial cells. This degradation process is facilitated by the hypermethylation of the Human antigen R (HuR) promoter, resulting in the transcriptional repression of HuR - a key regulator responsible for stabilizing p62 mRNA through direct binding. This hypermethylation is triggered by the activation of ribosomal protein S6, which is influenced by NNK exposure and subsequently amplifies the translation of DNA methyltransferase 3 alpha (DNMT3a). These findings provide crucial insights into the nature of p62 in both the development and potential treatment of tobacco-related lung cancer.
Collapse
Affiliation(s)
- Xuelei Liu
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325053, China
| | - Jingjing Wang
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Ziyi Yang
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325053, China
| | - Qipeng Xie
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325053, China; Department of Clinical Laboratory, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xinqi Diao
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325053, China
| | - Xiaoyan Yao
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325053, China
| | - Shirui Huang
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Ruifan Chen
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yunping Zhao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325053, China
| | - Tengda Li
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Minghua Jiang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325053, China; Department of Clinical Laboratory, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| | - Zhefeng Lou
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Chuanshu Huang
- Key Laboratory of Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325053, China.
| |
Collapse
|
48
|
Fu T, Yan P, Zhou L, Lu Z, Liu A, Ding X, Vannucci J, Hofman P, Swierniak A, Szurowska E, Zhang J, Li S. DeepGR: a deep-learning prognostic model based on glycolytic radiomics for non-small cell lung cancer. Transl Lung Cancer Res 2024; 13:2746-2760. [PMID: 39507025 PMCID: PMC11535831 DOI: 10.21037/tlcr-24-716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/25/2024] [Indexed: 11/08/2024]
Abstract
Background Glycolysis proved to have a prognostic value in lung cancer; however, to identify glycolysis-related genomic markers is expensive and challenging. This study aimed at identifying glycolysis-related computed tomography (CT) radiomics features to develop a deep-learning prognostic model for non-small cell lung cancer (NSCLC). Methods The study included 274 NSCLC patients from cohorts of The Second Affiliated Hospital of Soochow University (SZ; n=64), the Cancer Genome Atlas (TCGA)-NSCLC dataset (n=74), and the Gene Expression Omnibus dataset (n=136). Initially, the glycolysis enrichment scores were evaluated using a single-sample gene set enrichment analysis, and the cut-off values were optimized to investigate the prognostic potential of glycolysis genes. Radiomic features were then extracted using LIFEx software. The least absolute reduction and selection operator (LASSO) algorithm was employed to determine the glycolytic CT radiomics features. A deep-learning prognostic model was constructed by integrating CT radiomics and clinical features. The biological functions of the model were analyzed by incorporating RNA sequencing data. Results Kaplan-Meier curves indicated that elevated glycolysis levels were associated with poorer survival outcomes. The LASSO algorithm identified 11 radiomic features that were then selected for inclusion in the deep-learning model. They have shown significant discrimination capability in assessing glycolysis status, achieving an area under the curve value of 0.8442. The glycolysis-based radiomics deep-learning model was named the DeepGR model. This model was able to effectively predict the clinical outcomes of NSCLC patients with AUCs of 0.8760 and 0.8259 in the SZ and TCGA cohorts, respectively. High-risk DeepGR scores were strongly associated with poor overall survival, resting memory CD4+ T cells, and a high response to programmed cell death protein 1 immunotherapy. Conclusions The DeepGR model effectively predicted the prognosis of NSCLC patients.
Collapse
Affiliation(s)
- Tingting Fu
- Department of Radiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, China
| | - Peipei Yan
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Lina Zhou
- Health Management Center, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhihua Lu
- Department of Radiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, China
| | - Ao Liu
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiao Ding
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
| | - Jacopo Vannucci
- Thoracic Surgery Unit, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, FHU OncoAge, IHU RespirERA, Pasteur Hospital, BB-0033-00025, CHU Nice, University Côte d’Azur, Nice, France
| | - Andrzej Swierniak
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Edyta Szurowska
- 2nd Department of Radiology, Medical University of Gdansk, Gdansk, Poland
| | - Junjun Zhang
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Shicheng Li
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
49
|
Gao L, Wang W, Ma H, Yin M, Yang X, Han R, Ohara S, Kim D, Wang G. Bioinformatics analysis reveals SOD1 is a prognostic factor in lung adenocarcinoma. Transl Cancer Res 2024; 13:5522-5534. [PMID: 39525006 PMCID: PMC11543046 DOI: 10.21037/tcr-24-1400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/13/2024] [Indexed: 11/16/2024]
Abstract
Background Lung cancer is a major cause of cancer-related deaths worldwide. Unfortunately, non-small cell lung cancer (NSCLC) often lacks clear clinical symptoms and molecular markers for early diagnosis, which can hinder the initiation of timely treatments. In this study, we conducted an extensive bioinformatics analysis of copper-zinc superoxide dismutase (SOD1), a molecule linked to lung adenocarcinoma (LUAD) to enhance early detection and treatment approaches for this condition. Methods A bioinformatics analysis was conducted using a dataset from The Cancer Genome Atlas (TCGA) database. Several analytical methods, such as a differential expression analysis, a Kaplan-Meier survival analysis, a clinicopathological analysis, an enrichment analysis, protein-protein interaction (PPI) network construction using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database, and an immunoreactivity analysis of SOD1 expression in LUAD using TIMER were employed. We further validated the expression of SOD1 in LUAD through in vitro experiments using quantitative polymerase chain reaction (qPCR) and Western blot. Results Our findings indicate that LUAD tissues exhibited significantly higher expression levels of SOD1 than healthy tissues. The univariate Cox analysis showed that the elevated level was linked to unfavorable overall survival (OS) rates. Further, the Cox regression analysis of multiple variables suggested that elevated SOD1 expression levels acted as an autonomous prognosticator for unfavorable OS. We also conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, and a gene set enrichment analysis (GSEA) and observed differential pathway enrichment among patients with high SOD1 expression. In addition, a correlation between SOD1 and immune cell infiltration was found. The in vitro experiments confirmed that SOD1 expression was upregulated in LUAD. Conclusions SOD1 could serve as a reliable prognostic indicator in individuals diagnosed with LUAD. Our findings may prove valuable in the development of therapeutic and prognostic markers for LUAD. The potential clinical utility of SOD1 in LUAD requires further investigation.
Collapse
Affiliation(s)
- Ling Gao
- Clinical Laboratory, Chuxiong Yi Autonomous Prefecture People’s Hospital, Chuxiong, China
| | - Wei Wang
- Department of Neurosurgery, Chuxiong Yi Autonomous Prefecture People’s Hospital, Chuxiong, China
| | - Haishan Ma
- Clinical Laboratory, Chuxiong Yi Autonomous Prefecture People’s Hospital, Chuxiong, China
| | - Minghui Yin
- Clinical Laboratory, Chuxiong Yi Autonomous Prefecture People’s Hospital, Chuxiong, China
| | - Xuejiao Yang
- Clinical Laboratory, Chuxiong Yi Autonomous Prefecture People’s Hospital, Chuxiong, China
| | - Ruihui Han
- Clinical Laboratory, Chuxiong Yi Autonomous Prefecture People’s Hospital, Chuxiong, China
| | - Shuta Ohara
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Dohun Kim
- Department of Thoracic and Cardiovascular Surgery, College of Medicine, Chungbuk National University and Chungbuk National University Hospital, Cheongju, Korea
| | - Guangyan Wang
- Clinical Laboratory, Chuxiong Yi Autonomous Prefecture People’s Hospital, Chuxiong, China
| |
Collapse
|
50
|
Xu R, Han F, Zhao Y, Liu A, An N, Wang B, Zardo P, Sanz-Santos J, Franssen AJPM, de Loos ER, Zhao M. Role of CENPL, DARS2, and PAICS in determining the prognosis of patients with lung adenocarcinoma. Transl Lung Cancer Res 2024; 13:2729-2745. [PMID: 39507047 PMCID: PMC11535832 DOI: 10.21037/tlcr-24-696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/15/2024] [Indexed: 11/08/2024]
Abstract
Background Non-small cell lung cancer (NSCLC) accounts for about 85% of lung cancers, and is the leading cause of tumor-related death. Lung adenocarcinoma (LUAD) is the most prevalent subtype of NSCLC. Although significant progress of LUAD treatment has been made under multimodal strategies, the prognosis of advanced LUAD is still poor due to recurrence and metastasis. There is still a lack of reliable markers to evaluate the LUAD prognosis. This study aims to explore novel biomarkers and construct a prognostic model to predict the prognosis of LUAD patients. Methods The Genomic Data Commons-The Cancer Genome Atlas-Lung Adenocarcinoma (GDC-TCGA-LUAD) dataset was downloaded from the University of California, Santa Cruz (UCSC) Xena browser. The GSE72094 and GSE13213 datasets and corresponding clinical information were downloaded from the Gene Expression Omnibus (GEO) database. By analyzing these datasets using DESeq2 R package and Limma R package, differentially expressed genes (DEGs) were found. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were used to analyze possible enrichment pathways. A protein-protein interaction (PPI) network was constructed to explore possible relationship among DEGs by using the STRING database. A survival analysis was performed to identify reliable prognostic genes using the Kaplan-Meier method. A multi-omics analysis was performed using the Gene Set Cancer Analysis (GSCA). The Tumor Immune Estimation Score (TIMER) database was used to analyze the association between prognostic genes and immune infiltration. A Spearman correlation analysis was conducted to examine the correlation between prognostic genes and drug sensitivity. A multivariate Cox regression was used to identify independent prognostic factors. Next, a nomogram was constructed using the rms R package. Finally, the expressions of aspartyl-tRNA synthetase 2 (DARS2) and phosphoribosyl aminoimidazole carboxylase (PAICS) were detected using immunohistochemistry (IHC). Results We screened out 30 DEGs prior to functional enrichment and PPI network analysis revealing potential enrichment pathways and interactions of these DEGs. Then survival analysis revealed the CENPL, DARS2, and PAICS expression was negatively correlated with LUAD prognosis. Additionally, multi-omics analysis showed CENPL, DARS2, and PAICS expressions were significantly higher in LUAD tissues than normal tissues. CENPL, DARS2, and PAICS were all up-regulated in late stage and M1 stage. Correlation analysis indicated CENPL, DARS2, and PAICS may not be associated with activation or suppression of immune cells. Drug sensitivity analysis revealed many potentially effective drugs and small molecule compounds. Moreover, we successfully constructed a robust and stable nomogram by combining the DARS2 and PAICS expression with other clinicopathological variables. Finally, IHC results showed DARS2 and PAICS were significantly up-regulated in LUAD. Conclusions The CENPL, DARS2, and PAICS expression was negatively correlated with LUAD prognosis. A prognostic model, which integrated DARS2, PAICS, and other clinicopathological variables, was able to effectively predict LUAD patients prognosis.
Collapse
Affiliation(s)
- Rongjian Xu
- Department of Medical Microbiology, School of Basic Medicine, Qingdao University, Qingdao, China
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fengyi Han
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yandong Zhao
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ao Liu
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ning An
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Baogang Wang
- Department of Thoracic Surgery, The Anqiu Hospital of Traditional Chinese Medicine, Weifang, China
| | - Patrick Zardo
- Department of Cardiothoracic Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - José Sanz-Santos
- Pulmonology Department, Hospital Universitari Mútua Terrassa, University of Barcelona, Terrassa, Spain
| | - Aimée J. P. M. Franssen
- Division of General Thoracic Surgery, Department of Surgery, Zuyderland Medical Center, Heerlen, The Netherlands
| | - Erik R. de Loos
- Division of General Thoracic Surgery, Department of Surgery, Zuyderland Medical Center, Heerlen, The Netherlands
| | - Min Zhao
- Center of Laboratory Medicine, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| |
Collapse
|