1
|
Wang L, Pan X, Ye S, Huang Y, Wang M, Chen L, Zhou K, Han Y, Wu H. [ 18F]F-FAPI-42 PET dynamic imaging characteristics and multiparametric quantification of lung cancer: an exploratory study using uEXPLORER PET/CT. Eur J Nucl Med Mol Imaging 2025; 52:1685-1694. [PMID: 39760863 DOI: 10.1007/s00259-024-07064-3] [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: 10/31/2024] [Accepted: 12/29/2024] [Indexed: 01/07/2025]
Abstract
PURPOSE To explore the dynamic and parametric characteristics of [18F]F-FAPI-42 PET/CT in lung cancers. METHODS Nineteen participants with newly diagnosed lung cancer underwent 60-min dynamic [18F]F-FAPI-42 PET/CT. Time-activity curves (TAC) were generated for tumors and normal organs, with kinetic parameters (K1, K2, K3, K4, Ki) calculated. A new parameter, the K ratio (K1 + K3)/(K2 + K4), was introduced to measure net uptake efficiency. RESULTS In primary tumor (PT), [18F]F-FAPI-42 uptake showed a gradual increase followed by a plateau, contrasting with organs like the thyroid and pancreas, which showed rapid uptake and continuous washout. Compared to non-small cell lung cancer (NSCLC), small cell lung cancer (SCLC) lesions reached the plateau earlier (11 min vs. 14 min) but had a lower uptake. During the plateau phase, [18F]F-FAPI-42 demonstrated slight washout in SCLC, whereas its uptake increased slightly in NSCLC. Lymph node and distant metastases exhibited similar TAC profiles to primary tumors. Kinetic modeling revealed that an irreversible two-compartment model (irre-2TCM) best represented the pharmacokinetics of [18F]F-FAPI-42 in lung cancer, whereas re-2TCM was better suited for the pancreas and thyroid. Lower K1, K2, K3 and K4 were observed in PT compared to those in the pancreas and thyroid (P < 0.05), however, the K ratio in PT was found to be 2-3 times higher. SCLC had lower Ki and SUVmean than NSCLC (P < 0.05). Kinetic parameter differences were also observed between PT and metastatic lesions. Larger metastatic lymph nodes exhibited higher K1, Ki, and K ratio than smaller ones. CONCLUSION Lung cancers exhibit distinct [18F]F-FAPI-42 dynamic and kinetic characteristics compared to the thyroid gland and pancreas. Differences were also observed between SCLC and NSCLC, primary and metastatic lesions, as well as larger versus smaller lesions. These findings provide valuable insights into the in vivo pharmacokinetics of [18F]F-FAPI-42, potentially improving the diagnosis of lung cancer. TRIAL REGISTRATION ChiCTR2100045757. Registered April 24, 2021 retrospectively registered, http//www.chictr.org.cn.
Collapse
Affiliation(s)
- Lijuan Wang
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, China
- Department of Nuclear Medicine, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Xingzhu Pan
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, China
| | - Shimin Ye
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, China
| | - Yanchao Huang
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, China
| | - Meng Wang
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, China
| | - Li Chen
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, China
| | - Kemin Zhou
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, China
| | - Yanjiang Han
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, China
| | - Hubing Wu
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, China.
| |
Collapse
|
2
|
Martín F, Alcon C, Marín E, Morales-Sánchez P, Manzano-Muñoz A, Díaz S, García M, Samitier J, Lu A, Villanueva A, Reguart N, Teixido C, Montero J. Novel selective strategies targeting the BCL-2 family to enhance clinical efficacy in ALK-rearranged non-small cell lung cancer. Cell Death Dis 2025; 16:194. [PMID: 40113795 PMCID: PMC11926089 DOI: 10.1038/s41419-025-07513-3] [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: 02/21/2024] [Revised: 01/29/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
ALK (anaplastic lymphoma kinase) rearrangements represent the third most predominant driver oncogene in non-small cell lung cancer (NSCLC). Although ALK inhibitors are the tyrosine kinase inhibitors (TKIs) with the longest survival rates in lung cancer, the complex systemic clinical evaluation and the apoptotic cell death evasion of drug-tolerant persister (DTP) cancer cells may limit their therapeutic response. We found that dynamic BH3 profiling (DBP) presents an excellent predictive capacity to ALK-TKIs, that would facilitate their use in a clinical setting and complementing the readout of standard diagnostic assays. In addition, we revealed novel acute adaptive mechanisms in response to ALK inhibitors in cell lines and patient-derived tumor cells. Consistently, all our cell models confirmed a rapid downregulation of the sensitizer protein NOXA, leading to dependence on the anti-apoptotic protein MCL-1 after treatment with ALK-TKIs. In some cases, the anti-apoptotic protein BCL-xL may contribute equally to this anti-apoptotic response. Importantly, these acute dependencies could be prevented with BH3 mimetics in vitro and in vivo, blocking tumor adaptation to treatment. Finally, we also demonstrated how dual reactivation of PI3K/AKT and MAPK signaling pathways can impair lorlatinib response, which could be overcome with specific inhibitors of both signaling pathways. In conclusion, our findings propose several therapeutic combinations that should be explored in future clinical trials to enhance ALK inhibitors efficacy and improve the clinical response in a broad NSCLC patient population.
Collapse
Affiliation(s)
- Fernando Martín
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Clara Alcon
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Elba Marín
- Division of Medical Oncology, Hospital Clínic, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Unitat funcional de Tumors Toràcics, Hospital Clínic, Barcelona, Spain
| | - Paula Morales-Sánchez
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Albert Manzano-Muñoz
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Sherley Díaz
- Department of Pathology and CORE Molecular Biology Laboratory, Hospital Clínic, Barcelona, Spain
| | - Mireia García
- Department of Pathology and CORE Molecular Biology Laboratory, Hospital Clínic, Barcelona, Spain
| | - Josep Samitier
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
- Department of Electronics and Biomedical Engineering, Faculty of Physics, University of Barcelona, Barcelona, Spain
| | - Albert Lu
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Alberto Villanueva
- Chemoresistance and Predictive Factors Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
| | - Noemí Reguart
- Division of Medical Oncology, Hospital Clínic, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Unitat funcional de Tumors Toràcics, Hospital Clínic, Barcelona, Spain
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Cristina Teixido
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Unitat funcional de Tumors Toràcics, Hospital Clínic, Barcelona, Spain
- Department of Pathology and CORE Molecular Biology Laboratory, Hospital Clínic, Barcelona, Spain
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Joan Montero
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
3
|
Lin YD, Li HJ, Hong HZ, Qi YF, Li YY, Yang XN, Wu YL, Zhong WZ. Genomic profiling of aggressive pathologic features in lung adenocarcinoma. Lung Cancer 2025; 203:108460. [PMID: 40179539 DOI: 10.1016/j.lungcan.2025.108460] [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: 05/30/2024] [Revised: 02/02/2025] [Accepted: 02/26/2025] [Indexed: 04/05/2025]
Abstract
INTRODUCTION Pathologic features involving LVI (lympho-vascular invasion), PNI (perineural invasion), STAS (spread through air spaces), and Grade 3 pattern (from the International Association for the Study of Lung Cancer grading system) are related to having an aggressive phenotype and linked to poor prognosis. However, few studies have conducted in-depth analyses of these features simultaneously with genomic profiling. METHODS A total of 1559 sequencing of adenocarcinoma samples were included in the common driver mutations analysis, 1306 samples were brought into genomic mapping analysis. OncoSG's East Asian ancestry dataset was implemented for Tumor-Node-Metastasis-Biomarker (TNMB) classification and prognostic assessment. RESULTS EGFR was more significantly prevalent in LVI negativity (P = 0.021), STAS negativity (P = 0.002), and moderate grade (P < 0.001). ALK was significantly interrelated with LVI (P = 0.028), STAS (P < 0.001), and poor grade (P < 0.001); ROS1 and STAS positivity (P = 0.031), poor grade (P = 0.016) were significantly related. KRAS (P = 0.003) and BRAF-V600E (P = 0.002) were only significantly intertwined with poor grade. Apart from common driver mutations, TP53, CHEK2, KEAP1, PTEN, RB1, NF1 were significantly enriched in LVI samples (P < 0.05). TP53, PTEN, CTNNB1, HGF, NF1 were more prominent in STAS (P < 0.01). TP53, LRP1B, NF1 were significantly more prevalent in Grade 3 pattern (P < 0.001). The mixture of STK11, PTEN, and TOP2A generated by exclusive mutations may be a potential predictor of TNMB categorization towards survival. The HR of stage II compared I of TNMB was 2.28 (95 % CI 1.36-3.86, P < 0.001), while stage III compared II was 1.95 (95 % CI 1.04-3.21, P = 0.031). CONCLUSIONS This analysis demonstrated the correlation of pathologic features with common driver mutations, key mutations and canonical oncogenic signaling pathways. The data highlighted the similarities and differences among these features horizontally, and provide new insights in TNMB classification and prognostic assessment.
Collapse
Affiliation(s)
- Yi-Duo Lin
- School of Medicine, South China University of Technology, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hong-Ji Li
- School of Medicine, South China University of Technology, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hui-Zhao Hong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yi-Fan Qi
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yun-Yi Li
- School of Software Engineering, South China University of Technology, Guangzhou, China
| | - Xue-Ning Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Wen-Zhao Zhong
- School of Medicine, South China University of Technology, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
4
|
Wu S, Tu Q, Yuan H, Wu Z, Yang Y, Chen C, Huang C. Comprehensive Analysis for Predicting Prognoses and Immune Responses of m6A-Related lncRNAs in Women with Lung Adenocarcinoma. Biochem Genet 2024; 62:2702-2720. [PMID: 37999876 DOI: 10.1007/s10528-023-10572-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: 06/18/2023] [Accepted: 10/26/2023] [Indexed: 11/25/2023]
Abstract
During the past decade, the average 5-year survival rate of patients with Lung adenocarcinoma (LUAD) has remained < 20%, although the targeted therapies and novel immunotherapy approaches have held promise. Epigenetic modifications could provide prognostic value as molecular biomarkers, and we aimed to identify the independent risk of m6A-related lncRNAs to establish a risk model for the clinical prediction of prognoses in women with LUAD. In this study, we first assessed 31 N6-methyladenosine (m6A)-related lncRNAs associated with overall survival. Moreover, we evaluated the expression of the oncogenic driver and the tumor immune microenvironment (TIME) in two female LUAD subtypes (clusters 1 and 2) using consensus clustering. We also found 16 m6A-related lncRNAs as the independent prognostic indicator of women with LUAD and established a risk model developed from these lncRNAs. We comprehensively investigated the correlation between the TIME and m6A-related lncRNA and found that m6A-related lncRNA may significantly affect the immune cell infiltration level in LUAD. In conclusion, our study provides evidence on the prognostic prediction in women with LUAD and may help elucidate the processes and mechanisms of m6A-regulated lncRNAs.
Collapse
Affiliation(s)
- Sijie Wu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, #139 Renmin Road, Changsha, 410011, Hunan, China
| | - Qinxian Tu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, #139 Renmin Road, Changsha, 410011, Hunan, China
| | - Haoyong Yuan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, #139 Renmin Road, Changsha, 410011, Hunan, China
- Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, 410008, Hunan, China
| | - Zhongshi Wu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, #139 Renmin Road, Changsha, 410011, Hunan, China
- Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, 410008, Hunan, China
| | - Yifeng Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, #139 Renmin Road, Changsha, 410011, Hunan, China
| | - Chunyang Chen
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, #139 Renmin Road, Changsha, 410011, Hunan, China
- Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, 410008, Hunan, China
| | - Can Huang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, #139 Renmin Road, Changsha, 410011, Hunan, China.
- Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, 410008, Hunan, China.
| |
Collapse
|
5
|
Zaccagnino A, Vynnytska‐Myronovska B, Stöckle M, Junker K. Molecular and functional characterization of reversible-sunitinib-tolerance state in human renal cell carcinoma. J Cell Mol Med 2024; 28:e18329. [PMID: 38693863 PMCID: PMC11063727 DOI: 10.1111/jcmm.18329] [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: 01/22/2024] [Revised: 03/11/2024] [Accepted: 03/29/2024] [Indexed: 05/03/2024] Open
Abstract
Therapy failure with the tyrosine kinase inhibitor (TKI) sunitinib remains a great challenge in metastatic renal cell carcinoma (mRCC). Growing evidence indicates that the tumour subpopulation can enter a transient, non-mutagenic drug-tolerant state to endure the treatment underlying the minimal residual disease and tumour relapse. Drug tolerance to sunitinib remains largely unexplored in RCC. Here, we show that sunitinib-tolerant 786-O/S and Caki-2/S cells are induced by prolonged drug treatment showing reduced drug sensitivity, enhanced clonogenicity, and DNA synthesis. Sunitinib-tolerance developed via dynamic processes, including (i) engagement of c-MET and AXL pathways, (ii) alteration of stress-induced p38 kinase and pro-survival BCL-2 signalling, (iii) extensive actin remodelling, which was correlated with activation of focal adhesion proteins. Remarkably, the acute drug response in both sensitive and sunitinib-tolerant cell lines led to dramatic fine-tuning of the actin-cytoskeleton and boosted cellular migration and invasion, indicating that the drug-response might depend on cell state transition rather than pre-existing mutations. The drug-tolerant state was transiently acquired, as the cells resumed initial drug sensitivity after >10 passages under drug withdrawal, reinforcing the concept of dynamic regulation and phenotypic heterogeneity. Our study described molecular events contributing to the reversible switch into sunitinib-tolerance, providing possible novel therapeutic opportunities in RCC.
Collapse
Affiliation(s)
- Angela Zaccagnino
- Department of Urology and Pediatric UrologySaarland UniversityHomburgGermany
| | | | - Michael Stöckle
- Department of Urology and Pediatric UrologySaarland UniversityHomburgGermany
| | - Kerstin Junker
- Department of Urology and Pediatric UrologySaarland UniversityHomburgGermany
| |
Collapse
|
6
|
Huang Z, Xiao Z, Yu L, Liu J, Yang Y, Ouyang W. Tumor-associated macrophages in non-small-cell lung cancer: From treatment resistance mechanisms to therapeutic targets. Crit Rev Oncol Hematol 2024; 196:104284. [PMID: 38311012 DOI: 10.1016/j.critrevonc.2024.104284] [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: 10/28/2023] [Revised: 01/20/2024] [Accepted: 01/31/2024] [Indexed: 02/06/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) remains one of the leading causes of cancer-related deaths worldwide. Different treatment approaches are typically employed based on the stage of NSCLC. Common clinical treatment methods include surgical resection, drug therapy, and radiation therapy. However, with the introduction and utilization of immune checkpoint inhibitors, cancer treatment has entered a new era, completely revolutionizing the treatment landscape for various cancers and significantly improving overall patient survival. Concurrently, treatment resistance often poses a critical challenge, with many patients experiencing disease progression following an initial response due to treatment resistance. Increasing evidence suggests that the tumor microenvironment (TME) plays a pivotal role in treatment resistance. Tumor-associated macrophages (TAMs) within the TME can promote treatment resistance in NSCLC by secreting various cytokines activating signaling pathways, and interacting with other immune cells. Therefore, this article will focus on elucidating the key mechanisms of TAMs in treatment resistance and analyze how targeting TAMs can reduce the levels of treatment resistance in NSCLC, providing a comprehensive understanding of the principles and approaches to overcome treatment resistance in NSCLC.
Collapse
Affiliation(s)
- Zhenjun Huang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Ziqi Xiao
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Liqing Yu
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Jiayu Liu
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Yihan Yang
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang 330006, Jiangxi Province, China.
| | - Wenhao Ouyang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| |
Collapse
|
7
|
Chrisochoidou Y, Roy R, Farahmand P, Gonzalez G, Doig J, Krasny L, Rimmer EF, Willis AE, MacFarlane M, Huang PH, Carragher NO, Munro AF, Murphy DJ, Veselkov K, Seckl MJ, Moffatt MF, Cookson WOC, Pardo OE. Crosstalk with lung fibroblasts shapes the growth and therapeutic response of mesothelioma cells. Cell Death Dis 2023; 14:725. [PMID: 37938546 PMCID: PMC10632403 DOI: 10.1038/s41419-023-06240-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: 06/18/2023] [Revised: 10/12/2023] [Accepted: 10/20/2023] [Indexed: 11/09/2023]
Abstract
Mesothelioma is an aggressive cancer of the mesothelial layer associated with an extensive fibrotic response. The latter is in large part mediated by cancer-associated fibroblasts which mediate tumour progression and poor prognosis. However, understanding of the crosstalk between cancer cells and fibroblasts in this disease is mostly lacking. Here, using co-cultures of patient-derived mesothelioma cell lines and lung fibroblasts, we demonstrate that fibroblast activation is a self-propagated process producing a fibrotic extracellular matrix (ECM) and triggering drug resistance in mesothelioma cells. Following characterisation of mesothelioma cells/fibroblasts signalling crosstalk, we identify several FDA-approved targeted therapies as far more potent than standard-of-care Cisplatin/Pemetrexed in ECM-embedded co-culture spheroid models. In particular, the SRC family kinase inhibitor, Saracatinib, extends overall survival well beyond standard-of-care in a mesothelioma genetically-engineered mouse model. In short, we lay the foundation for the rational design of novel therapeutic strategies targeting mesothelioma/fibroblast communication for the treatment of mesothelioma patients.
Collapse
Affiliation(s)
| | - Rajat Roy
- Division of Cancer, Imperial College, Du Cane Road, London, W12 0NN, UK
| | - Pooyeh Farahmand
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Guadalupe Gonzalez
- Department of Computing, Faculty of Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Jennifer Doig
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Lukas Krasny
- Molecular and Systems Oncology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Ella F Rimmer
- Division of Cancer, Imperial College, Du Cane Road, London, W12 0NN, UK
| | - Anne E Willis
- MRC Toxicology Unit, Tennis Ct Rd, Cambridge, CB2 1QR, UK
| | | | - Paul H Huang
- Molecular and Systems Oncology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Neil O Carragher
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XR, UK
| | - Alison F Munro
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XR, UK
| | - Daniel J Murphy
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Kirill Veselkov
- Division of Cancer, Imperial College, Du Cane Road, London, W12 0NN, UK
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Michael J Seckl
- Division of Cancer, Imperial College, Du Cane Road, London, W12 0NN, UK
| | - Miriam F Moffatt
- National Heart and Lung Institute, Imperial College, Dovehouse St, London, SW3 6LY, UK
| | - William O C Cookson
- National Heart and Lung Institute, Imperial College, Dovehouse St, London, SW3 6LY, UK.
| | - Olivier E Pardo
- Division of Cancer, Imperial College, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
8
|
Ilesanmi A, Dairo G, Salimat S, Bodun DS, Awoyale B, Balogun TA. Identification of bioactive compounds from Vaccinium vitis-idaea L. (Lingonberry) as inhibitors for treating KRAS-associated cancer: a computational approach. In Silico Pharmacol 2023; 11:32. [PMID: 37915613 PMCID: PMC10616029 DOI: 10.1007/s40203-023-00165-1] [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: 07/29/2023] [Accepted: 10/04/2023] [Indexed: 11/03/2023] Open
Abstract
Lung cancer is the cancer of the lung's epithelial cells typically characterized by difficulty breathing, chest pain, blood-stained coughs, headache, and weight loss. If left unmanaged, lung cancer can spread to other body parts. While several treatment methods exist for managing lung cancer, exploring natural plant sources for developing therapeutics offers great potential in complementing other treatment approaches. In this study, we evaluated the bioactive compounds in Vaccinium vitis-idaea for treating KRAS-associated lung cancer types. In this study, we concentrated on inhibiting the mutated Kirsten rat sarcoma viral oncogene homolog (KRAS) by targeting an associated protein (Phosphodiesterase 6δ) to which KRAS form complexes. We evaluated bioactive compounds from Lingonberry (Vaccinium vitis-idaea L.), adopting computational approaches such as molecular docking, molecular dynamics simulation, molecular mechanics/generalized Born surface area (MM/GBSA) calculations, and pharmacokinetics analysis. A total of 26 out of 39 bioactive compounds of Vaccinium vitis-idaea L. had a higher binding affinity to the target receptor than an approved drug, Sotorasib. Also, further analyses of all lead/top compounds in this study identified (+)-Catechin (Cianidanol), Arbutin, Resveratrol, and Sinapic acid, to be potential drug candidates that could be pursued. In sum, Arbutin, (+)-Catechin, and Sinapic acid are predicted to be the top compound of Vaccinium vitis-idaea L. because of their pharmacokinetic properties and drug-likeness attributes. Also, their stability to the target receptor makes them a potential drug candidate that could be explored for treating KRAS mutation-associated lung cancer. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-023-00165-1.
Collapse
Affiliation(s)
- Ayooluwa Ilesanmi
- Department of Sciences and Mathematics, Mississippi University for Women, Columbus, USA
| | - Gbenga Dairo
- Department of Biological Sciences, Western Illinois University, Macomb, IL USA
| | - Sofela Salimat
- Department of Chemistry, University of Lagos, Lagos, Nigeria
| | - Damilola S. Bodun
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| | - Bibiire Awoyale
- Department of Chemistry, University of Ilorin, Ilorin, Nigeria
| | - Toheeb A. Balogun
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
- Department of Biological Sciences, University of California, San Diego, La Jolla, USA
| |
Collapse
|
9
|
Song X, Lan Y, Zheng X, Zhu Q, Liao X, Liu K, Zhang W, Peng Q, Zhu Y, Zhao L, Chen X, Shu Y, Yang K, Hu J. Targeting drug-tolerant cells: A promising strategy for overcoming acquired drug resistance in cancer cells. MedComm (Beijing) 2023; 4:e342. [PMID: 37638338 PMCID: PMC10449058 DOI: 10.1002/mco2.342] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 08/29/2023] Open
Abstract
Drug resistance remains the greatest challenge in improving outcomes for cancer patients who receive chemotherapy and targeted therapy. Surmounting evidence suggests that a subpopulation of cancer cells could escape intense selective drug treatment by entering a drug-tolerant state without genetic variations. These drug-tolerant cells (DTCs) are characterized with a slow proliferation rate and a reversible phenotype. They reside in the tumor region and may serve as a reservoir for resistant phenotypes. The survival of DTCs is regulated by epigenetic modifications, transcriptional regulation, mRNA translation remodeling, metabolic changes, antiapoptosis, interactions with the tumor microenvironment, and activation of signaling pathways. Thus, targeting the regulators of DTCs opens a new avenue for the treatment of therapy-resistant tumors. In this review, we first provide an overview of common characteristics of DTCs and the regulating networks in DTCs development. We also discuss the potential therapeutic opportunities to target DTCs. Last, we discuss the current challenges and prospects of the DTC-targeting approach to overcome acquired drug resistance. Reviewing the latest developments in DTC research could be essential in discovering of methods to eliminate DTCs, which may represent a novel therapeutic strategy for preventing drug resistance in the future.
Collapse
Affiliation(s)
- Xiaohai Song
- Department of General SurgeryGastric Cancer CenterLaboratory of Gastric CancerState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Yang Lan
- Department of General SurgeryGastric Cancer CenterLaboratory of Gastric CancerState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Xiuli Zheng
- Department of RadiologyHuaxi MR Research Center (HMRRC) and Critical Care MedicinePrecision Medicine Center, Frontiers Science Center for Disease‐Related Molecular Network, West China HospitalSichuan UniversityChengduChina
| | - Qianyu Zhu
- Department of General SurgeryGastric Cancer CenterLaboratory of Gastric CancerState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Xuliang Liao
- Department of General SurgeryGastric Cancer CenterLaboratory of Gastric CancerState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Kai Liu
- Department of General SurgeryGastric Cancer CenterLaboratory of Gastric CancerState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Weihan Zhang
- Department of General SurgeryGastric Cancer CenterLaboratory of Gastric CancerState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - QiangBo Peng
- Department of General SurgeryGastric Cancer CenterLaboratory of Gastric CancerState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Yunfeng Zhu
- Department of General SurgeryGastric Cancer CenterLaboratory of Gastric CancerState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Linyong Zhao
- Department of General SurgeryGastric Cancer CenterLaboratory of Gastric CancerState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Xiaolong Chen
- Department of General SurgeryGastric Cancer CenterLaboratory of Gastric CancerState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Yang Shu
- Department of General SurgeryGastric Cancer CenterLaboratory of Gastric CancerState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Kun Yang
- Department of General SurgeryGastric Cancer CenterLaboratory of Gastric CancerState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Jiankun Hu
- Department of General SurgeryGastric Cancer CenterLaboratory of Gastric CancerState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
10
|
Chen S, Lin J, Zhao J, Lin Q, Liu J, Wang Q, Mui R, Ma L. FBXW7 attenuates tumor drug resistance and enhances the efficacy of immunotherapy. Front Oncol 2023; 13:1147239. [PMID: 36998461 PMCID: PMC10043335 DOI: 10.3389/fonc.2023.1147239] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/24/2023] [Indexed: 03/17/2023] Open
Abstract
FBXW7 (F-box and WD repeat domain containing 7) is a critical subunit of the Skp1-Cullin1-F-box protein (SCF), acting as an E3 ubiquitin ligase by ubiquitinating targeted protein. Through degradation of its substrates, FBXW7 plays a pivotal role in drug resistance in tumor cells and shows the potential to rescue the sensitivity of cancer cells to drug treatment. This explains why patients with higher FBXW7 levels exhibit higher survival times and more favorable prognosis. Furthermore, FBXW7 has been demonstrated to enhance the efficacy of immunotherapy by targeting the degradation of specific proteins, as compared to the inactivated form of FBXW7. Additionally, other F-box proteins have also shown the ability to conquer drug resistance in certain cancers. Overall, this review aims to explore the function of FBXW7 and its specific effects on drug resistance in cancer cells.
Collapse
Affiliation(s)
- Shimin Chen
- Department of Oncology, Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jichun Lin
- Department of Oncology, Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jiaojiao Zhao
- Department of Oncology, Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Qian Lin
- Department of Oncology, Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jia Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - Qiang Wang
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Ryan Mui
- Department of Gastroenterology, Sparrow Hospital, Lansing, MI, United States
| | - Leina Ma
- Department of Oncology, Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
- *Correspondence: Leina Ma,
| |
Collapse
|
11
|
Kleczko EK, Poczobutt JM, Navarro AC, Laskowski J, Johnson AM, Korpela SP, Gurule NJ, Heasley LE, Hopp K, Weiser-Evans MC, Gottlin EB, Bushey RT, Campa MJ, Patz EF, Thurman JM, Nemenoff RA. Upregulation of complement proteins in lung cancer cells mediates tumor progression. Front Oncol 2023; 12:1045690. [PMID: 36686777 PMCID: PMC9849673 DOI: 10.3389/fonc.2022.1045690] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/02/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction In vivo, cancer cells respond to signals from the tumor microenvironment resulting in changes in expression of proteins that promote tumor progression and suppress anti-tumor immunity. This study employed an orthotopic immunocompetent model of lung cancer to define pathways that are altered in cancer cells recovered from tumors compared to cells grown in culture. Methods Studies used four murine cell lines implanted into the lungs of syngeneic mice. Cancer cells were recovered using FACS, and transcriptional changes compared to cells grown in culture were determined by RNA-seq. Results Changes in interferon response, antigen presentation and cytokine signaling were observed in all tumors. In addition, we observed induction of the complement pathway. We previously demonstrated that activation of complement is critical for tumor progression in this model. Complement can play both a pro-tumorigenic role through production of anaphylatoxins, and an anti-tumorigenic role by promoting complement-mediated cell killing of cancer cells. While complement proteins are produced by the liver, expression of complement proteins by cancer cells has been described. Silencing cancer cell-specific C3 inhibited tumor growth In vivo. We hypothesized that induction of complement regulatory proteins was critical for blocking the anti-tumor effects of complement activation. Silencing complement regulatory proteins also inhibited tumor growth, with different regulatory proteins acting in a cell-specific manner. Discussion Based on these data we propose that localized induction of complement in cancer cells is a common feature of lung tumors that promotes tumor progression, with induction of complement regulatory proteins protecting cells from complement mediated-cell killing.
Collapse
Affiliation(s)
- Emily K. Kleczko
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Joanna M. Poczobutt
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Andre C. Navarro
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jennifer Laskowski
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Amber M. Johnson
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Sean P. Korpela
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Natalia J. Gurule
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Lynn E. Heasley
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Katharina Hopp
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Mary C.M. Weiser-Evans
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Elizabeth B. Gottlin
- Department of Radiology, Duke University School of Medicine, Durham, NC, United States
| | - Ryan T. Bushey
- Department of Radiology, Duke University School of Medicine, Durham, NC, United States
| | - Michael J. Campa
- Department of Radiology, Duke University School of Medicine, Durham, NC, United States
| | - Edward F. Patz
- Department of Radiology, Duke University School of Medicine, Durham, NC, United States
- Department of Pharmacology and Cancer Biology, Duke School of Medicine, Durham, NC, United States
| | - Joshua M. Thurman
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Raphael A. Nemenoff
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
12
|
Sobreira da Silva MJ, Serpa Osorio-de-Castro CG, Paes RD, Negrete CL, Eugênio E, Moraes EL, Livinalli A. Potential interactions between antineoplastic agents and medicines used to treat Covid-19. J Oncol Pharm Pract 2022; 28:1737-1748. [PMID: 34637360 PMCID: PMC9619075 DOI: 10.1177/10781552211040494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Cancer patients with Covid-19 are exposed to treatment combinations that can potentially result in interactions that adversely affect patient outcomes. This study aimed to identify potential drug-drug interactions between antineoplastic agents and medicines used to treat Covid-19. METHODS We conducted a search for potential interactions between 201 antineoplastic agents and 26 medicines used to treat Covid-19 on the Lexicomp® and Micromedex® databases. The following data were extracted: interaction severity ("major" and "contraindicated") and interaction effects (pharmacokinetic and pharmacodynamic). We also sought to identify the therapeutic indication of the antineoplastic drugs involved in the potential drug-drug interactions. RESULTS A total of 388 "major" or "contraindicated" drug-drug interactions were detected. Eight drugs or combinations (baricitinib, lopinavir/ritonavir, atazanavir, darunavir, azithromycin, chloroquine, hydroxychloroquine, and sirolimus) accounted for 91.5% of these interactions. The class of antineoplastic agents with the greatest potential for interaction was tyrosine kinase inhibitors (accounting for 46.4% of all interactions). The findings show that atazanavir, baricitinib, and lopinavir/ritonavir can affect the treatment of all common types of cancer. The most common pharmacokinetic effect of the potential drug-drug interactions was increased plasma concentration of the antineoplastic medicine (39.4%). CONCLUSIONS Covid-19 is a recent disease and pharmacological interventions are undergoing constant modification. This study identified a considerable number of potential drug-drug interactions. In view of the vulnerability of patients with cancer, it is vital that health professionals carefully assess the risks and benefits of drug combinations.
Collapse
Affiliation(s)
- Mario Jorge Sobreira da Silva
- National Cancer Institute of
Brazil, Brazil,Mario Jorge Sobreira da Silva, Rua Marquês
de Pombal, 125, 3° andar – Centro, Rio de Janeiro, RJ 20230-240, Brazil.
| | | | | | | | | | | | | |
Collapse
|
13
|
Rare EGFR E709-T710delinsX: Molecular characteristics and superior response to afatinib treatment in NSCLC patients. Lung Cancer 2022; 172:117-123. [DOI: 10.1016/j.lungcan.2022.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 11/19/2022]
|
14
|
Das R, Jakubowski MA, Spildener J, Cheng YW. Identification of Novel MET Exon 14 Skipping Variants in Non-Small Cell Lung Cancer Patients: A Prototype Workflow Involving in Silico Prediction and RT-PCR. Cancers (Basel) 2022; 14:cancers14194814. [PMID: 36230737 PMCID: PMC9563401 DOI: 10.3390/cancers14194814] [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: 06/30/2022] [Revised: 09/11/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Background and aims: The MET exon 14 skipping (METex14) is an oncogenic driver mutation that provides a therapeutic opportunity in non-small cell lung cancer (NSCLCs) patients. This event often results from sequence changes at the MET canonical splicing sites. We characterize two novel non-canonical splicing site variants of MET that produce METex14. Materials and Methods: Two variants were identified in three advanced-stage NSCLC patients in a next-generation sequencing panel. The potential impact on splicing was predicted using in silico tools. METex14 mutation was confirmed using reverse transcription (RT)-PCR and a Sanger sequencing analysis on RNA extracted from stained cytology smears. Results: The interrogated MET (RefSeq ID NM_000245.3) variants include a single nucleotide substitution, c.3028+3A>T, in intron 14 and a deletion mutation, c.3012_3028del, in exon 14. The in silico prediction analysis exhibited reduced splicing strength in both variants compared with the MET normal transcript. The RT-PCR and subsequent Sanger sequencing analyses confirmed METex14 skipping in all three patients carrying these variants. Conclusion: This study reveals two non-canonical MET splice variants that cause exon 14 skipping, concurrently also proposes a clinical workflow for the classification of such non-canonical splicing site variants detected by routine DNA-based NGS test. It shows the usefulness of in silico prediction to identify potential METex14 driver mutation and exemplifies the opportunity of routine cytology slides for RNA-based testing.
Collapse
Affiliation(s)
| | | | | | - Yu-Wei Cheng
- Correspondence: ; Tel.: +1-216-445-0757; Fax: +1-216-445-0681
| |
Collapse
|
15
|
Hata T, Sakaguchi C, Hirano K, Kobe H, Ishida M, Nakano T, Tachibana Y, Tamiya N, Shiotsu S, Takeda T, Yamada T, Yokoyama T, Tsuchiya M, Nagasaka Y. Efficacy and safety of immuno-chemotherapy in patients with advanced non-small-cell lung cancer harboring oncogenic mutations: a multicenter retrospective study. J Cancer Res Clin Oncol 2022; 149:2475-2482. [PMID: 35737092 DOI: 10.1007/s00432-022-04125-8] [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: 05/02/2022] [Accepted: 06/07/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE The effect of immuno-chemotherapy on patients with advanced non-small-cell lung cancer (NSCLC) harboring oncogenic mutations remains poorly understood. This study aimed to characterize the efficacy of immuno-chemotherapy and determine the optimal treatment strategy for such patients. METHODS We conducted this retrospective cohort study on patients with NSCLC harboring oncogenic driver alterations and treated with an immune checkpoint inhibitor combined with chemotherapy at five institutions. The clinical characteristics and outcomes of immuno-chemotherapy for NSCLC with oncogenic mutations in a real-world setting were analyzed. RESULTS Among 846 patients diagnosed with advanced or recurrent NSCLC between April 2017 and April 2021, 43 patients with oncogenic mutations were treated with immuno-chemotherapy. The median age of patients was 68 (range 44-78) years; 42% of patients never smoked, and adenocarcinoma was the most common histology (95%). In patients with KRAS mutations (n = 10) or PD-L1 expression of 50% or greater (n = 10), the disease control rate was 100%. The median progression-free survival (PFS) was 5.4, 6.3, and 8.9 months in patients harboring mutations in EGFR, KRAS, and other genes, respectively (P = 0.22). Patients with PD-L1 expression of 50% or greater had significantly longer median PFS than patients with PD-L1 expression of less than 50% (16.4 vs. 5.1 months; P = 0.001). Two patients experienced grade 3 immuno-related adverse events. CONCLUSION Immuno-chemotherapy has a clinical benefit and is safe for patients with oncogenic mutations. Notably, patients with PD-L1 expression of 50% or more experience greater benefit from immuno-chemotherapy than those with PD-L1 expression of less than 50%.
Collapse
Affiliation(s)
- Tae Hata
- Department of Respiratory Medicine, Rakuwakai Otowa Hospital, 2 Otowachinji-cho, Yamashina, Kyoto, 607-8062, Japan.
| | - Chikara Sakaguchi
- Department of Medical Oncology, Rakuwakai Otowa Hospital, Kyoto, Japan
| | - Keita Hirano
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroshi Kobe
- Department of Respiratory Medicine, Kurashiki Central Hospital, Okayama, Japan
| | - Masaki Ishida
- Department of Respiratory Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takayuki Nakano
- Department of Respiratory Medicine, Japanese Red Cross Kyoto Daini Hospital, Kyoto, Japan
| | - Yusuke Tachibana
- Department of Respiratory Medicine, Japanese Red Cross Kyoto Daiichi Hospital, Kyoto, Japan
| | - Nobuyo Tamiya
- Department of Respiratory Medicine, Rakuwakai Otowa Hospital, 2 Otowachinji-cho, Yamashina, Kyoto, 607-8062, Japan
| | - Shinsuke Shiotsu
- Department of Respiratory Medicine, Japanese Red Cross Kyoto Daiichi Hospital, Kyoto, Japan
| | - Takayuki Takeda
- Department of Respiratory Medicine, Japanese Red Cross Kyoto Daini Hospital, Kyoto, Japan
| | - Tadaaki Yamada
- Department of Respiratory Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshihide Yokoyama
- Department of Respiratory Medicine, Kurashiki Central Hospital, Okayama, Japan
| | - Michiko Tsuchiya
- Department of Respiratory Medicine, Rakuwakai Otowa Hospital, 2 Otowachinji-cho, Yamashina, Kyoto, 607-8062, Japan
| | - Yukio Nagasaka
- Department of Respiratory Medicine, Rakuwakai Otowa Hospital, 2 Otowachinji-cho, Yamashina, Kyoto, 607-8062, Japan
| |
Collapse
|
16
|
Nie Z, Chen M, Gao Y, Huang D, Cao H, Peng Y, Guo N, Wang F, Zhang S. Ferroptosis and Tumor Drug Resistance: Current Status and Major Challenges. Front Pharmacol 2022; 13:879317. [PMID: 35668934 PMCID: PMC9163417 DOI: 10.3389/fphar.2022.879317] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Ferroptosis is a novel type of regulated cell death, whose unique metabolic characteristics are commonly used to evaluate the conditions of various diseases especially in tumors. Accumulating evidence supports that ferroptosis can regulate tumor development, metastasis, and therapeutic responses. Considering to the important role of chemotherapy in tumor treatment, drug resistance has become the most serious challenge. Revealing the molecular mechanism of ferroptosis is expected to solve tumor drug resistance and find new therapies to treat cancers. In this review, we discuss the relationship between ferroptosis and tumor drug resistance, summarize the abnormal ferroptosis in tissues of different cancer types and current research progress and challenges in overcoming treatment resistance, and explore the concept of targeting ferroptosis to improve tumor treatment outcomes.
Collapse
Affiliation(s)
- Zhenyu Nie
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Mei Chen
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Yuanhui Gao
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Denggao Huang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Hui Cao
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Yanling Peng
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Na Guo
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Fei Wang
- Department of Urology, Hainan General Hospital, Affiliated Hainan Hospital of Hainan Medical University, Haikou, China
- *Correspondence: Shufang Zhang, ; Fei Wang,
| | - Shufang Zhang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
- *Correspondence: Shufang Zhang, ; Fei Wang,
| |
Collapse
|
17
|
Bai Y, Pei Y, Xia L, Ma L, Deng S. A Novel Immune-Prognosis Index Predicts the Benefit of Lung Adenocarcinoma Patients. Front Pharmacol 2022; 13:818170. [PMID: 35614936 PMCID: PMC9124834 DOI: 10.3389/fphar.2022.818170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 04/08/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Constructed an immune-prognosis index (IPI) and divided lung adenocarcinoma (LUAD) patients into different subgroups according to IPI score, describe the molecular and immune characteristics of patients between different IPI subgroups, and explore their response to immune checkpoint blockade (ICB) treatment. Methods: Based on the transcriptome profile of LUAD patients in TCGA and immune gene sets from ImmPort and InnateDB, 15 hub immune genes were identified through correlation and Bayesian causal network analysis. Then, IPI was constructed with 5 immune genes by using COX regression analysis and verified with external datasets (GSE30219, GSE37745, GSE68465, GSE126044 and GSE135222). Finally, the characteristics and the response to ICB treatment of LUAD patients between two different IPI subgroups were analyzed. Results: IPI was constructed based on the expression of 5 genes, including A2M, ADRB1, ADRB2, VIPR1 and PTH1R. IPI-high LUAD patients have a better overall survival than IPI-low LUAD patients, consistent with the results in the GEO cohorts. The comprehensive results showed that patients in the IPI-high subgroup were exhibited characters as metabolism-related signaling pathways activation, lower TP53 and TTN mutation rate, more infiltrations of CD8 T cells, dendritic cells and macrophages M1, especially earned more benefit from ICB treatment. In contrast, patients in the IPI-low subgroup were exhibited characters as p53 signaling pathways activation, higher TP53 and TTN mutation rate, more infiltrations of resting memory CD4 T cells, macrophages M2, immune-suppressive response and less benefit from ICB treatment. Conclusion: IPI is a potentially valuable prognostic evaluation method for LUAD, which works well in the benefit predicting of LUAD patients within ICB treatment.
Collapse
Affiliation(s)
- Yuquan Bai
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yun Pei
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China
| | - Liang Xia
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Ma
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Senyi Deng
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Senyi Deng,
| |
Collapse
|
18
|
Zhu G, Chi H, Liu M, Yin Y, Diao H, Liu Z, Guo Z, Xu W, Xu J, Cui C, Xing XJ, Ma K. Multifunctional "ball-rod" Janus nanoparticles boosting Fenton reaction for ferroptosis therapy of non-small cell lung cancer. J Colloid Interface Sci 2022; 621:12-23. [PMID: 35447518 DOI: 10.1016/j.jcis.2022.04.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/02/2022] [Accepted: 04/04/2022] [Indexed: 01/13/2023]
Abstract
Ferroptosis is a newly found cell death mechanism, which could bypass apoptosis and reverse multidrug resistance of tumors. However, efficient induction of tumor ferroptosis remains a challenge. In this study, multifunctional "ball-rod" Janus nanoparticles (FTG/L&SMD) were constructed for non-small cell lung cancer (NSCLC) ferroptosis treatment. Protected by tannic acid (TA), FTG/L&SMD maintains long-term function in blood circulation, while modification by 2, 3-dimethylmaleic anhydride (DMMA) confers the FTG/L&SMD with pH-responsive charge reversal. Glucose oxidase (GOD) on FTG/L&SMD catalyzes glucose to produce H2O2. Then, iron ion converts H2O2 to highly active hydroxyl radicals (OH•) via Fenton reaction, leading to lethal lipid peroxidation (LPO) accumulation. Meanwhile, TA reduces Fe3+ to Fe2+ to boost Fenton reaction cycle. Sor down-regulated glutathione peroxidase 4 (GPX4) expression in another pathway to induce ferroptosis synergistically. In vitro studies have shown that compared with sorafenib (Sor), FTG/L&SMD not only has more efficient tumor targeting and higher cytotoxicity, but also inhibits tumor migration. In vivo antitumor therapy experiments demonstrate that FTG/L&SMD inhibits tumor growth efficiently, and its toxicity is negligible. In general, FTG/L&SMD can initiate Fenton reaction cycle and reinforced ferroptosis to kill tumor cells, which is a promising anti-tumor nano-drug for NSCLC.
Collapse
Affiliation(s)
- Guang Zhu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Hao Chi
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Meichen Liu
- Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Yalin Yin
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - He Diao
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Zicheng Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Zhaoming Guo
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Weiping Xu
- School of Ocean Science and Technology, Dalian University of Technology, Panjin 124221, China
| | - Jianqiang Xu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China; Panjin Institute of Industrial Technology, Dalian University of Technology, Panjin 124221, China
| | - Changhao Cui
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China.
| | - Xiao-Jin Xing
- Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China.
| | - Kun Ma
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China.
| |
Collapse
|
19
|
Li K, Liu J, Wu L, Xiao Y, Li J, Du H, Zhao Z, Sun C, Zhao Y, Yang J, Wu D, Zhao Z, Chen B. Genomic correlates of programmed cell death ligand 1 (PD-L1) expression in Chinese lung adenocarcinoma patients. Cancer Cell Int 2022; 22:138. [PMID: 35346207 PMCID: PMC8962080 DOI: 10.1186/s12935-022-02488-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/24/2022] [Indexed: 12/26/2022] Open
Abstract
Abstract
Background
Although PD-L1 expression is a crucial predictive biomarker for immunotherapy, it can be influenced by many factors.
Methods
A total of 248 Chinese patients with lung adenocarcinoma was retrospectively identified. Data for clinical features, gene alternations, signaling pathways and immune signatures was analyzed among negative expression group (TPS < 1%, n = 124), intermediate expression group (1% ≤ TPS < 50%, n = 93), and high expression group (TPS ≥ 50%, n = 38). Clinical outcomes among different expression groups were also evaluated from public database.
Results
Firstly, high tumor mutation burden was significantly associated with high PD-L1 expression in these Chinese patients with lung adenocarcinoma. In addition, gene alternations including TP53, PRKDC, KMT2D, TET1 and SETD2 apparently occurred in high PD-L1 expression group. Moreover, pathway analysis showed that mutations involving in DDR pathway, TP53 pathway, cell-cycle pathway and NOTCH pathway were obviously varied among three PD-L1 expression groups. Besides, most of patients in high PD-L1 expression group from TCGA database were determined as high-grade immune subtypes (C2-C4), showing significant higher proportions of IFN-gamma, CD8+ T-cells, NK cells, NK CD56 dim cells, Th1 cells, Th2 cells (P < 0.0001). Moreover, SETD2 mutation slightly correlated with overall survival from MSKCC cohort (HR 1.92 [95%CI 0.90–4.10], P = 0.085), and the percentage of IFN-gamma was significantly higher in SETD2 mutant group than in wild-type group (P < 0.01).
Conclusions
This study illustrated in-depth genomic correlates of PD-L1 expression in Chinese lung adenocarcinoma patients and relevant immune signatures from public database, which might interpret more potential molecular mechanisms for immunotherapy in NSCLC.
Collapse
|
20
|
Liu T, Guo L, Liu G, Hu X, Li X, Zhang J, Dai Z, Yu P, Jiang M, Wang J, Zhang J. Molecular Characterization of the Clinical and Tumor Immune Microenvironment Signature of 5-methylcytosine-Related Regulators in non-small Cell Lung Cancer. Front Cell Dev Biol 2021; 9:779367. [PMID: 34858994 PMCID: PMC8632062 DOI: 10.3389/fcell.2021.779367] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 10/19/2021] [Indexed: 12/28/2022] Open
Abstract
Background: DNA methylation is an important epigenetic modification, among which 5-methylcytosine methylation (5mC) is generally associated with tumorigenesis. Nonetheless, the potential roles of 5mC regulators in the tumor microenvironment (TME) remain unclear. Methods: The 5mC modification patterns of 1,374 lung adenocarcinoma samples were analyzed systematically. The correlation between the 5mC modification and tumor microenvironment cell infiltration was further assessed. The 5mCscore was developed to evaluate tumor mutation burden, immune check-point inhibitor response, and the clinical prognosis of individual tumors. Results: Three 5mC modification patterns were established based on the clinical characteristics of 21 5mC regulators. According to the differential expression of 5mC regulators, three distinct 5mC gene cluster were also identified, which showed distinct TME immune cell infiltration patterns and clinical prognoses. The 5mCscore was constructed to evaluate the tumor mutation burden, immune check-point inhibitor response, and prognosis characteristics. We found that patients with a low 5mCscore had significant immune cell infiltration and increased clinical benefit. Conclusion: This study indicated that the 5mC modification is involved in regulating TME infiltration remodeling. Targeting 5mC modification regulators might be a novel strategy to treat lung cancer.
Collapse
Affiliation(s)
- Taisheng Liu
- Department of Thoracic Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Liyi Guo
- The Sixth People's Hospital of Huizhou City, Huiyang Hospital Affiliated to Southern Medical University, Huizhou, China
| | - Guihong Liu
- Department of Radiation Oncology, DongGuan Tungwah Hospital, Dongguan, China
| | - Xiaoshan Hu
- Department of Internal Medicine of Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Xiaoning Li
- Department of Thoracic Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Jinye Zhang
- Department of Thoracic Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Zili Dai
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou, China
| | - Peng Yu
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou, China
| | - Ming Jiang
- Department of Breast Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Jian Wang
- Department of Thoracic Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Jian Zhang
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou, China
| |
Collapse
|
21
|
Guo Y, Song Q, Jiang M, Guo Y, Xu P, Zhang Y, Fu CC, Fang Q, Zeng M, Yao X. Histological Subtypes Classification of Lung Cancers on CT Images Using 3D Deep Learning and Radiomics. Acad Radiol 2021; 28:e258-e266. [PMID: 32622740 DOI: 10.1016/j.acra.2020.06.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 06/05/2020] [Accepted: 06/05/2020] [Indexed: 12/24/2022]
Abstract
RATIONALE AND OBJECTIVES Histological subtypes of lung cancers are critical for clinical treatment decision. In this study, we attempt to use 3D deep learning and radiomics methods to automatically distinguish lung adenocarcinomas (ADC), squamous cell carcinomas (SCC), and small cell lung cancers (SCLC) respectively on Computed Tomography images, and then compare their performance. MATERIALS AND METHODS 920 patients (mean age 61.2, range, 17-87; 340 Female and 580 Male) with lung cancer, including 554 patients with ADC, 175 patients with lung SCC and 191 patients with SCLC, were included in this retrospective study from January 2013 to August 2018. Histopathologic analysis was available for every patient. The classification models based on 3D deep learning (named the ProNet) and radiomics (named com_radNet) were designed to classify lung cancers into the three types mentioned above according to histopathologic results. The training, validation and testing cohorts counted 0.70, 0.15, and 0.15 of the whole datasets respectively. RESULTS The ProNet model used to classify the three types of lung cancers achieved the F1-scores of 90.0%, 72.4%, 83.7% in ADC, SCC, and SCLC respectively, and the weighted average F1-score of 73.2%. For com_radNet, the F1-scores achieved 83.1%, 75.4%, 85.1% in ADC, SCC, and SCLC, and the weighted average F1-score was 72.2%. The area under the receiver operating characteristic curve of the ProNet model and com_radNet were 0.840 and 0.789, and the accuracy were 71.6% and 74.7% respectively. CONCLUSION The ProNet and com_radNet models we developed can achieve high performance in distinguishing ADC, SCC, and SCLC and may be promising approaches for non-invasive predicting histological subtypes of lung cancers.
Collapse
|
22
|
Cabanos HF, Hata AN. Emerging Insights into Targeted Therapy-Tolerant Persister Cells in Cancer. Cancers (Basel) 2021; 13:cancers13112666. [PMID: 34071428 PMCID: PMC8198243 DOI: 10.3390/cancers13112666] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/19/2021] [Accepted: 05/19/2021] [Indexed: 12/25/2022] Open
Abstract
Drug resistance is perhaps the greatest challenge in improving outcomes for cancer patients undergoing treatment with targeted therapies. It is becoming clear that "persisters," a subpopulation of drug-tolerant cells found in cancer populations, play a critical role in the development of drug resistance. Persisters are able to maintain viability under therapy but are typically slow cycling or dormant. These cells do not harbor classic drug resistance driver alterations, and their partial resistance phenotype is transient and reversible upon removal of the drug. In the clinic, the persister state most closely corresponds to minimal residual disease from which relapse can occur if treatment is discontinued or if acquired drug resistance develops in response to continuous therapy. Thus, eliminating persister cells will be crucial to improve outcomes for cancer patients. Using lung cancer targeted therapies as a primary paradigm, this review will give an overview of the characteristics of drug-tolerant persister cells, mechanisms associated with drug tolerance, and potential therapeutic opportunities to target this persister cell population in tumors.
Collapse
Affiliation(s)
- Heidie Frisco Cabanos
- Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA;
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Aaron N. Hata
- Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA;
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Correspondence: ; Tel.: +1-617-724-3442
| |
Collapse
|
23
|
Luo Z, Zhang H, Xiao Y, Wang R, Zhang L, Huang C, Cao Y, Sun C, Zhao Y, Lin H, Wu D, Wang T. Durable Response to Immunotherapy With Antiangiogenic Drug in Large-Cell Lung Carcinoma With Multiple Fulminant Postoperative Metastases: A Case Report. Front Oncol 2021; 11:633446. [PMID: 34094914 PMCID: PMC8173040 DOI: 10.3389/fonc.2021.633446] [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: 11/25/2020] [Accepted: 04/19/2021] [Indexed: 11/13/2022] Open
Abstract
Immunotherapy alone or chemo-immunotherapy has recently been recommended for treating advanced lung carcinoma in patients without driver mutations. However, the efficacy of immunotherapy and molecular mechanism in large-cell lung cancer (LCLC) remains unclear. Here, we reported a rare case of multiple fulminant postoperative body and mouth metastases in LCLC treating with combination immunotherapy. Initially, the patient was diagnosed as early stage LCLC and underwent a radical resection of the right lower lobe. Just one month later, multiple fulminant body and mouth lesions appeared in the right upper arm, right elbow, right waist, and tongue root. Meanwhile, serum neuron specific enolase (NSE) concentration dramatically increased from 12.12 to 30.14 ng/ml. Immumohistochemistry findings demonstrated moderate PD-L1 expressions with tumor proportion score (TPS), while next-generation sequencing indicated moderate tumor mutational burden (TMB) levels and gene mutations in PBRM1 L1230P and TP53 L194R of both foci. Besides, loss of heterozygosity (LOH) at human leukocyte antigen (HLA) class I (HLA-A*02:03, HLA-B*55:02 and HLA-C*12:03) were detected in the right upper arm metastasis, which may facilitate malignant postoperative metastases in this case. Notably, this patient received combination therapy with anti-PD-1 antibody sintilimab plus anlotinib, and achieved a partial response for at least 12 months. Using an integrated computational method, the mutant peptide TEIPENDIPL derived from PBRM1 L1230P was predicted to be a specific neoantigen and could still be presented by HLA-B*40:01. This case suggests that immunotherapy plus antiangiogenic drug may provide an alternative therapeutic option for advanced LCLC patients without common gene mutations.
Collapse
Affiliation(s)
- Zhilin Luo
- Department of Thoracic Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Zhang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yajie Xiao
- Department of Medicine, YuceBio Technology Co. Ltd., Shenzhen, China
| | - Rui Wang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liping Zhang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chenglu Huang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Cao
- Department of Thoracic Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chao Sun
- Department of Medicine, YuceBio Technology Co. Ltd., Shenzhen, China
| | - Yongtian Zhao
- Department of Medicine, YuceBio Technology Co. Ltd., Shenzhen, China
| | - Hanqing Lin
- Department of Medicine, YuceBio Technology Co. Ltd., Shenzhen, China
| | - Dongfang Wu
- Department of Medicine, YuceBio Technology Co. Ltd., Shenzhen, China
| | - Tianhu Wang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
24
|
Zhou Q, Chen W, Fan Z, Chen Z, Liang J, Zeng G, Liu L, Liu W, Yang T, Cao X, Yu B, Xu M, Chen YG, Chen L. Targeting hyperactive TGFBR2 for treating MYOCD deficient lung cancer. Theranostics 2021; 11:6592-6606. [PMID: 33995678 PMCID: PMC8120205 DOI: 10.7150/thno.59816] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/23/2021] [Indexed: 01/16/2023] Open
Abstract
Purpose: Clinical success of cancer therapy is severely limited by drug resistance, attributed in large part to the loss of function of tumor suppressor genes (TSGs). Developing effective strategies to treat those tumors is challenging, but urgently needed in clinic. Experimental Design: MYOCD is a clinically relevant TSG in lung cancer patients. Our in vitro and in vivo data confirm its tumor suppressive function. Further analysis reveals that MYOCD potently inhibits stemness of lung cancer stem cells. Mechanistically, MYOCD localizes to TGFBR2 promoter region and thereby recruits PRMT5/MEP50 complex to epigenetically silence its transcription. Conclusions: NSCLC cells deficient of MYOCD are particularly sensitive to TGFBR kinase inhibitor (TGFBRi). TGFBRi and stemness inhibitor synergize with existing drugs to treat MYOCD deficient lung cancers. Our current work shows that loss of function of MYOCD creates Achilles' heels in lung cancer cells, which might be exploited in clinic.
Collapse
|
25
|
Socinski MA, Pennell NA, Davies KD. MET Exon 14 Skipping Mutations in Non-Small-Cell Lung Cancer: An Overview of Biology, Clinical Outcomes, and Testing Considerations. JCO Precis Oncol 2021; 5:PO.20.00516. [PMID: 34036238 PMCID: PMC8140815 DOI: 10.1200/po.20.00516] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/19/2021] [Accepted: 03/04/2021] [Indexed: 12/11/2022] Open
Affiliation(s)
| | - Nathan A. Pennell
- Department of Hematology and Medical Oncology, Cleveland Clinic, Cleveland, OH
| | - Kurtis D. Davies
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
26
|
Gmeiner WH. Recent Advances in Our Knowledge of mCRC Tumor Biology and Genetics: A Focus on Targeted Therapy Development. Onco Targets Ther 2021; 14:2121-2130. [PMID: 33790575 PMCID: PMC8007558 DOI: 10.2147/ott.s242224] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/11/2021] [Indexed: 12/20/2022] Open
Abstract
Metastatic colorectal cancer (mCRC) remains a highly lethal malignancy although considerable progress has resulted from characterizing molecular alterations such as RAS mutation status and extent of microsatellite instability (MSI) to guide optimal use of available therapies. The availability of gene expression profiling, next generation sequencing technologies, proteomics analysis and other technologies provides high resolution information on individual tumors, including metastatic lesions to better define intra-tumor and inter-tumor heterogeneity. Recent literature applying this information to further customize personalized therapies is reviewed. Current biomarker-based stratification used to select optimal therapy that is personalized to the mutation profile of individual tumors is described. Recent literature using whole exome sequencing of metastatic lesions and primary CRC tumors and other advanced technologies to more fully elucidate the tumor biology specific to mCRC sub-types and to develop more precise therapies that improve outcomes is also reviewed.
Collapse
Affiliation(s)
- William H Gmeiner
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
27
|
De Toma A, Lo Russo G, Signorelli D, Pagani F, Randon G, Galli G, Prelaj A, Ferrara R, Proto C, Ganzinelli M, Zilembo N, de Braud F, Garassino MC. Uncommon targets in non-small cell lung cancer: Everyone wants a slice of cake. Crit Rev Oncol Hematol 2021; 160:103299. [PMID: 33722699 DOI: 10.1016/j.critrevonc.2021.103299] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/14/2020] [Accepted: 03/10/2021] [Indexed: 01/15/2023] Open
Abstract
Target therapies completely changed the clinical approach in EGFR mutated and ALK rearranged non-small cell lung cancer, ensuring these patients exceptional outcomes with a better toxicity profile compared to conventional chemotherapy. In recent years, beyond EGFR and ALK alterations, new data are emerging about less common alterations, new drugs have been already approved and others agents have been recently investigated or are currently under investigation. In this review we will discuss some uncommon alterations in non-small cell lung cancer such as ROS1, BRAF, RET, HER2, NTRK, MET and other targets that are in an early evaluation phase. We will summarize the characteristics of patients harboring these alterations, the already approved or under investigation therapies and the related resistance mechanisms.
Collapse
Affiliation(s)
- Alessandro De Toma
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - Giuseppe Lo Russo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Diego Signorelli
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo Pagani
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giovanni Randon
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giulia Galli
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Arsela Prelaj
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Department of Electronics, Information, and Bioengineering, Polytechnic University of Milan, Milan, Italy
| | - Roberto Ferrara
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Claudia Proto
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Monica Ganzinelli
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Nicoletta Zilembo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marina Chiara Garassino
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
28
|
Yu Y, Qian J, Shen L, Ji W, Lu S. Distinct profile of cell-free DNA in malignant pleural effusion of non-small cell lung cancer and its impact on clinical genetic testing. Int J Med Sci 2021; 18:1510-1518. [PMID: 33628109 PMCID: PMC7893565 DOI: 10.7150/ijms.52306] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/18/2020] [Indexed: 02/06/2023] Open
Abstract
Cell-free DNA (cfDNA) in supernatant of pleural effusion from advanced NSCLC patients has been proved as surrogate sample detecting therapeutic targets as well as tumor mutation burden (TMB). As recently reported, cfDNA in pleural effusion supernatant is superior to plasma in TMB evaluation. It is reasonable to hypothesize that cfDNA profile in pleural effusion (PE) and plasma might be different. It remains to be elucidated why cfDNA in PE supernatant impacts on genetic analysis. Consequently, the approach dealing with cfDNA from PE supernatant might need to be different from that for plasma cfDNA in order to obtain accurate clinical genetic testing result. Methods: Pleural effusion samples from 32 patients with stage IV lung adenocarcinoma were collected. Supernatant and sediment were processed separately to extract Cell-free DNA as well as sediment DNA (PE-S). cfDNA from pleural effusion was analyzed by Agilent 2100 bioanalyzer. Libraries were prepared by 1) direct use of the total cfDNA without fragmentation step (PE-FL) or 2) use of full-length cfDNA fragmented to 150-250bp (PE-F), 3) use of cfDNA fragments enriched to ~167bp (PE-E167) as well as 4) use of cfDNA fragments larger than 500bp enriched (PE-E500). All samples were subjected to targeted next-generation sequencing (NGS) with a panel of 448 cancer-related genes as well as a panel of 10 NSCLC driver genes. Results: cfDNA were successfully extracted from 30 MPE samples. cfDNA displayed distinct profile in supernatant of malignant pleural effusion from that of plasma cfDNA. No statistical difference in detection of hotspot variations between PE-E167 and PE-F by 448-gene or 10-gene panel. While TMB from PE-F samples was significantly higher than that from PE-E167 and PE-FL. Higher TMB from PE-F was resulted from cancer-unspecific variants with low allele frequency (0.1%-1%) which were mainly introduced by long-fragment cfDNA. Similar genetic profile was observed between paired cfDNA of PE-FL and cfDNA of PE-E167. Conclusion: Long-fragment cfDNA in the PE supernatant will introduce low abundant cancer unrelated variants which leads overestimation of TMB. Paired PE-FL and PE-E167 gave comparable outcomes. Direct use of the total cfDNA without fragmentation step (PE-FL) is recommended for library preparation of NGS testing in clinical practice to exclude interference from long fragments of the cfDNA.
Collapse
Affiliation(s)
| | | | | | | | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
29
|
Li T, Li J. [Efficacy of Immune Checkpoint Inhibitors for Non-small Cell Lung Cancer
with Rare Mutation]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2021; 24:19-24. [PMID: 33478186 PMCID: PMC7849033 DOI: 10.3779/j.issn.1009-3419.2020.102.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
驱动基因的发现及针对驱动基因的靶向治疗已显著提高了肺癌患者的生存质量和时间,但目前对于BRAF、HER2、MET、RET等少见驱动基因改变肺癌患者的靶向药物的选择仍然较少。近年来免疫检查点抑制剂在肺癌治疗中取得了一定的疗效,但因为少见驱动基因突变的肺癌患者本身样本量少,开展大规模临床随机对照试验尚存在一定的困难,目前此类患者接受免疫检查点抑制剂治疗的疗效情况仍不明确。本文将对目前已掌握的免疫检查点抑制剂治疗BRAF、HER2、MET、RET等少见驱动基因改变肺癌患者的临床研究结果进行综述,以期在一定程度上为临床工作提供一些依据和参考。
Collapse
Affiliation(s)
- Teng Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences
and Peking Union Medical College, Beijing 100021, China
| | - Junling Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences
and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
30
|
Chen X, Gao A, Zhang F, Yang Z, Wang S, Fang Y, Li J, Wang J, Shi W, Wang L, Zheng Y, Sun Y. ILT4 inhibition prevents TAM- and dysfunctional T cell-mediated immunosuppression and enhances the efficacy of anti-PD-L1 therapy in NSCLC with EGFR activation. Theranostics 2021; 11:3392-3416. [PMID: 33537094 PMCID: PMC7847666 DOI: 10.7150/thno.52435] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/24/2020] [Indexed: 12/13/2022] Open
Abstract
Rationale: Immune checkpoint inhibitors (ICIs) against the PD-1/PD-L1 pathway showed limited success in non-small cell lung cancer (NSCLC) patients, especially in those with activating epidermal growth factor receptor (EGFR) mutations. Elucidation of the mechanisms underlying EGFR-mediated tumor immune escape and the development of effective immune therapeutics are urgently needed. Immunoglobulin-like transcript (ILT) 4, a crucial immunosuppressive molecule initially identified in myeloid cells, is enriched in solid tumor cells and promotes the malignant behavior of NSCLC. However, the upstream regulation of ILT4 overexpression and its function in tumor immunity of NSCLC with EGFR activation remains unclear. Methods: ILT4 expression and EGFR phosphorylation in human NSCLC tissues and cell lines were analyzed using immunohistochemistry (IHC), real-time PCR, Western blotting, immunofluorescence, and flow cytometry. The molecular signaling for EGFR-regulated ILT4 expression was investigated using mRNA microarray and The Cancer Genome Atlas (TCGA) database analyses and then confirmed by Western blotting. The regulation of tumor cell proliferation and apoptosis by ILT4 was examined by CCK8 proliferation and apoptosis assays. The impact of ILT4 and PD-L1 on tumor-associated macrophage (TAM) recruitment and polarization was evaluated using Transwell migration assay, flow cytometry, enzyme linked immunosorbent assay (ELISA) and real-time PCR, while their impact on T cell survival and cytotoxicity was analyzed by CFSE proliferation assay, apoptotic assay, flow cytometry, ELISA and cytolytic assay. Tumor immunotherapy models targeting at paired Ig-like receptor B (PIR-B, an ortholog of ILT4 in mouse)/ILT4 and/or PD-L1 were established in C57BL/6 mice inoculated with stable EGFR- overexpressing Lewis lung carcinoma (LLC) cells and in humanized NSG mice inoculated with EGFR mutant, gefitinib-resistant PC9 (PC9-GR) or EGFR-overexpressing wild type H1299 cells. PIR-B and ILT4 inhibition was implemented by infection of specific knockdown lentivirus and PD-L1 was blocked using human/mouse neutralizing antibodies. The tumor growth model was established in NSG mice injected with PIR-B-downregulated LLC cells to evaluate the effect of PIR-B on tumor proliferation. The frequencies and phenotypes of macrophages and T cells in mouse spleens and blood were detected by flow cytometry while those in tumor tissues were determined by IHC and immunofluorescence. Results: We found that ILT4 expression in tumor cells was positively correlated with EGFR phosphorylation in human NSCLC tissues. Using NSCLC cell lines, we demonstrated that ILT4 was upregulated by both tyrosine kinase mutation-induced and epidermal growth factor (EGF)-dependent EGFR activation and subsequent AKT/ERK1/2 phosphorylation. Overexpressed ILT4 in EGFR-activated tumor cells induced TAM recruitment and M2-like polarization, which impaired T cell function. ILT4 also directly inhibited T cell proliferation, cytotoxicity, and IFN-γ expression and secretion. In EGFR-activated cell lines in vitro and in wild-type EGFR-activated C57BL/6 and humanized NSG immunotherapy models in vivo, either ILT4 (PIR-B) or PD-L1 inhibition enhanced anti-tumor immunity and suppressed tumor progression by counteracting TAM- and dysfunctional T cell- induced immuno-suppressive TME; the combined inhibition of both molecules showed the most dramatic tumor retraction. Surprisingly, in EGFR mutant, TKI resistant humanized NSG immunotherapy model, ILT4 inhibition alone rather than in combination with a PD-L1 inhibitor suppressed tumor growth and immune evasion. Conclusions: ILT4 was induced by activation of EGFR-AKT and ERK1/2 signaling in NSCLC cells. Overexpressed ILT4 suppressed tumor immunity by recruiting M2-like TAMs and impairing T cell response, while ILT4 inhibition prevented immunosuppression and tumor promotion. Furthermore, ILT4 inhibition enhanced the efficacy of PD-L1 inhibitor in EGFR wild-type but not in EGFR mutant NSCLC. Our study identified novel mechanisms for EGFR-mediated tumor immune escape, and provided promising immunotherapeutic strategies for patients with EGFR-activated NSCLC.
Collapse
|
31
|
Tabbò F, Passiglia F, Novello S. Upfront Management of ALK-Rearranged Metastatic Non-small Cell Lung Cancer: One Inhibitor Fits All? Curr Oncol Rep 2021; 23:10. [PMID: 33387080 DOI: 10.1007/s11912-020-00989-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2020] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW Anaplastic lymphoma kinase (ALK) rearrangements represent a seldom event in non-small cell lung cancer (NSCLC). Given the oncogene alteration, ALK targeting represents the main therapeutic strategy. Here, we review evidence regarding ALK inhibitors (ALKi): clinical activity, safety profiles, financial costs, and biomarkers of efficacy. RECENT FINDINGS During the past 10 years, multiple ALKi have been developed, and four different compounds are currently available as upfront options for ALK+ NSCLC patients: crizotinib, ceritinib, alectinib, and brigatinib. Second-generation (2G) ALKi demonstrated superior clinical activity in terms of median progression-free survival (mPFS), objective response rate (ORR), intracranial disease control, and duration of response (DOR) when compared with crizotinib. 2G ALKi represent the current gold-standard first-line treatment for ALK-rearranged metastatic NSCLC. Among all available options, in our opinion, alectinib has likely the best profile of clinical activity and safety, thus emerging as the best upfront therapy. More insights will come from ongoing trials and analysis of biomarkers.
Collapse
Affiliation(s)
- Fabrizio Tabbò
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, TO, Italy
| | - Francesco Passiglia
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, TO, Italy
| | - Silvia Novello
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, TO, Italy.
| |
Collapse
|
32
|
Bai Y, Deng S. A six-long noncoding RNA model predicts prognosis in lung adenocarcinoma. Transl Cancer Res 2020; 9:7505-7518. [PMID: 35117351 PMCID: PMC8799207 DOI: 10.21037/tcr-20-2436] [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: 06/29/2020] [Accepted: 10/21/2020] [Indexed: 02/05/2023]
Abstract
Background The incidence and mortality of lung cancer rank first among various malignant tumors. The lack of clear molecular classification and effective individualized treatment greatly limits the treatment benefits of patients. Long non-coding RNAs (lncRNAs) have been demonstrated widely involve in tumor progressing, and been proved easy to detect for occupying majority in transcriptome. However, less work focuses on studying the potency of lncRNAs as molecular typing and prognostic indicator in lung cancer. Methods Based on the 448 lung adenocarcinoma (LUAD) samples and the expression of 14,127 lncRNAs from the Cancer Genome Atlas (TCGA) database, we constructed a co-expression network using weighted gene co-expression network analysis. Then based on the feature module and the overall survival of patients, we constructed a risk score model through Cox proportional hazards regression and verified it with a validation cohort. Finally, according to the median of risk score, the function of this model was enriched. Results We identified a module containing 123 lncRNAs that is related with the prognosis of LUAD. Using univariate and multivariate Cox proportional hazards regression with lasso regression, six lncRNAs were identified to construct a risk score model. The calculation formula shown as follows: risk score = (−0.3057 × EXPVIM-AS1) + (0.9678 × EXPAC092811.1) + (1.0829 × EXPNFIA-AS1) + (−0.3505 × EXPAL035701.1) + (3.9378 × EXPAC079336.4) + (−0.2810 × EXPAL121790.2). Six-lncRNA model can be used as an independent prognostic indicator in LUAD (P<0.001) and the area under the 5-year receiver operating characteristic (ROC) curve is 0.715. Conclusions We developed a six-lncRNA model, which could be used for predicting prognosis and guiding medical treatment in LUAD patients.
Collapse
Affiliation(s)
- Yuquan Bai
- Department of Thoracic Surgery research laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Senyi Deng
- Department of Thoracic Surgery research laboratory, West China Hospital, Sichuan University, Chengdu, China.,Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| |
Collapse
|
33
|
In Vitro and In Silico Evaluation of Anticancer Activity of New Indole-Based 1,3,4-Oxadiazoles as EGFR and COX-2 Inhibitors. Molecules 2020; 25:molecules25215190. [PMID: 33171861 PMCID: PMC7664637 DOI: 10.3390/molecules25215190] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) and cyclooxygenase-2 (COX-2) are crucial targetable enzymes in cancer management. Therefore, herein, new 2-[(5-((1H-indol-3-yl)methyl)-1,3,4-oxadiazol-2-yl)thio]-N-(thiazol/benzothiazol-2-yl)acetamides (2a-i) were designed and synthesized as EGFR and COX-2 inhibitors. The cytotoxic effects of compounds 2a-i on HCT116 human colorectal carcinoma, A549 human lung adenocarcinoma, and A375 human melanoma cell lines were determined using MTT assay. 2-[(5-((1H-Indol-3-yl)methyl)-1,3,4-oxadiazol-2-yl)thio]-N-(6-ethoxybenzothiazol-2-yl)acetamide (2e) exhibited the most significant anticancer activity against HCT116, A549, and A375 cell lines with IC50 values of 6.43 ± 0.72 μM, 9.62 ± 1.14 μM, and 8.07 ± 1.36 μM, respectively, when compared with erlotinib (IC50 = 17.86 ± 3.22 μM, 19.41 ± 2.38 μM, and 23.81 ± 4.17 μM, respectively). Further mechanistic assays demonstrated that compound 2e enhanced apoptosis (28.35%) in HCT116 cells more significantly than erlotinib (7.42%) and caused notable EGFR inhibition with an IC50 value of 2.80 ± 0.52 μM when compared with erlotinib (IC50 = 0.04 ± 0.01 μM). However, compound 2e did not cause any significant COX-2 inhibition, indicating that this compound showed COX-independent anticancer activity. The molecular docking study of compound 2e emphasized that the benzothiazole ring of this compound occupied the allosteric pocket in the EGFR active site. In conclusion, compound 2e is a promising EGFR inhibitor that warrants further clinical investigations.
Collapse
|
34
|
Li J, Gao A, Zhang F, Wang S, Wang J, Wang J, Han S, Yang Z, Chen X, Fang Y, Jiang G, Sun Y. ILT3 promotes tumor cell motility and angiogenesis in non-small cell lung cancer. Cancer Lett 2020; 501:263-276. [PMID: 33152402 DOI: 10.1016/j.canlet.2020.10.048] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/24/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022]
Abstract
Immunoglobulin-like transcript (ILT) 3 is an immunosuppressive molecule that negatively regulates myeloid cell activation. ILT3 overexpression in tumor cells induces immune escape of solid tumors and facilitates invasion of monocytic acute myeloid leukemia cells. However, the expression and function of ILT3 in non-small cell lung cancer (NSCLC) cells remain elusive. Herein, we found that ILT3 was enriched in human NSCLC cells, and predicted advanced disease and poor overall survival. ILT3 overexpression enhanced the migration and invasion of NSCLC cells and tubule formation of human umbilical vein endothelial cells by upregulating and interacting with its ligand apolipoprotein E (ApoE) in vitro. Mechanistically, ILT3 recruited SHP2 and SHIP1, and subsequently activated ERK1/2 signaling mediating epithelial-mesenchymal transition (EMT) and increasing vascular endothelial growth factor (VEGF)-A expression in NSCLC cells, which are responsible for tumor cell motility and angiogenesis, respectively. Using murine metastasis models, we further confirmed ILT3 promoted NSCLC metastasis and explored the exact correlation of ILT3 with ApoE, EMT, and VEGF-A in vivo. These results unraveled novel mechanisms for ILT3-induced tumor progression and proposed ILT3 as a potential therapeutic target and prognostic biomarker for NSCLC patients.
Collapse
Affiliation(s)
- Juan Li
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, PR China
| | - Aiqin Gao
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, PR China
| | - Fang Zhang
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, PR China
| | - Shuyun Wang
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, PR China
| | - Jingnan Wang
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, PR China
| | - Jing Wang
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, PR China
| | - Shuyi Han
- Medical Research and Laboratory Diagnostic Center, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, PR China
| | - Zijiang Yang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xiaozheng Chen
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yuying Fang
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, PR China
| | - Guosheng Jiang
- Department of Immunology, College of Basic Medical, Binzhou Medical University, Yantai, Shandong, 256600, PR China
| | - Yuping Sun
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, PR China; Department of Oncology, Jinan Central Hospital affiliated to Shandong First Medical University, Jinan, Shandong, 250013, PR China.
| |
Collapse
|
35
|
Chen Z, Fan Z, Dou X, Zhou Q, Zeng G, Liu L, Chen W, Lan R, Liu W, Ru G, Yu L, He QY, Chen L. Inactivation of tumor suppressor gene Clusterin leads to hyperactivation of TAK1-NF-κB signaling axis in lung cancer cells and denotes a therapeutic opportunity. Am J Cancer Res 2020; 10:11520-11534. [PMID: 33052230 PMCID: PMC7545994 DOI: 10.7150/thno.44829] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 09/04/2020] [Indexed: 12/31/2022] Open
Abstract
Purpose: Clinical success of precision medicine is severely limited by de novo or acquired drug resistance. It remains a clinically unmet need to treat these patients. Tumor suppressor genes (TSGs) play a critical role in tumorigenesis and impact the therapeutic effect of various treatments. Experimental Design: Using clinical data, in vitro cell line data and in vivo mouse model data, we revealed the tumor suppressive role of Clusterin in lung cancer. We also delineated the signaling cascade elicited by loss of function of CLU in NSCLC cells and tested precision medicine for CLU deficient lung cancers. Results: CLU is a potent and clinically relevant TSG in lung cancer. Mechanistically, CLU inhibits TGFBR1 to recruit TRAF6/TAB2/TAK1 complex and thus inhibits activation of TAK1- NF-κB signaling axis. Lung cancer cells with loss of function of CLU show exquisite sensitivity to TAK1 inhibitors. Importantly, we show that a significant portion of Kras mutation positive NSCLC patients are concurrently deficient of CLU and that TAK1 kinase inhibitor synergizes with existing drugs to treat this portion of lung cancers patients. Conclusions: Combinational treatment with TAK1 inhibitor and MEK1/2 inhibitor effectively shrank Kras mutation positive and CLU deficient NSCLC tumors. Moreover, we put forward a concept that loss of function of a TSG rewires signaling network and thereby creates an Achilles' heel in tumor cells which could be exploited in precision medicine.
Collapse
|
36
|
Thistlethwaite PA. The sparkle of creativity. J Thorac Cardiovasc Surg 2020; 160:740-752. [PMID: 32800264 PMCID: PMC8865752 DOI: 10.1016/j.jtcvs.2020.03.176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/03/2020] [Accepted: 03/14/2020] [Indexed: 01/27/2023]
Abstract
Thinking creatively leads to new discoveries and better ways to do things, enhances personal well-being and self-worth, and ultimately benefits our patients.
Collapse
|
37
|
Xu J, Qu S, Sun N, Zhang W, Zhang J, Song Q, Lin M, Gao W, Zheng Q, Han M, Na C, Xu R, Chang X, Yang X, Huang J. Construction of a reference material panel for detecting KRAS/ NRAS/ EGFR/ BRAF/ MET mutations in plasma ctDNA. J Clin Pathol 2020; 74:314-320. [PMID: 32817175 PMCID: PMC8070650 DOI: 10.1136/jclinpath-2020-206745] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/28/2020] [Accepted: 07/02/2020] [Indexed: 12/24/2022]
Abstract
Background The absence of high-quality next-generation sequencing (NGS) reference material (RM) has impeded the clinical use of liquid biopsies with plasma cell-free DNA (cfDNA) in China. Objective This study aimed to develop a national RM panel for external quality assessment and performance evaluation during kit registration of non-small-cell lung cancer (NSCLC)-related Kirsten rat sarcoma viral oncogene (KRAS)/neuroblastoma ras oncogene (NRAS)/epidermal growth factor receptor (EGFR)/B-type Raf kinase (BRAF)/mesenchymal–epithelial transition factor (MET) genetic assays using plasma circulating tumor DNA (ctDNA). Methods Mutation cell lines detected by NGS and validated by Sanger sequencing were selected to establish the RM. Cell line genomic DNA was sheared and used to spike basal plasma cfDNA at 10% concentration. Then, the calibration accuracy was determined by four sequencing platforms. Average values were adopted and diluted to 0.1%, 0.3%, 1% and 3% concentrations with basal plasma as the RM panel. Then, five manufacturers were invited to evaluate the performance of the RM panel. Results 20 cell lines with 23 clinically important mutations were selected, including six mutations in KRAS, two mutations in NRAS, three in BRAF, four in phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), six in EGFR, one EGFR Gain (4-5 copy) and one MET Gain (2-5 copy). The RM panel consisted of 87 samples, including these 21 mutations at four concentrations (0.1%, 0.3%, 1% and 3%), one MET gain, one EGFR gain and one wild type. The detection rate was 100% for the 3%, 1% and 0.3% samples at all five companies. For the 0.1% concentration, 15 samples had inconsistent results, but at least three companies had correct results for each mutation. Conclusion RM for a KRAS/NRAS/EGFR/BRAF/MET mutation panel for plasma ctDNA was developed, which will be essential for quality control of the performance of independent laboratories.
Collapse
Affiliation(s)
- Jun Xu
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Shoufang Qu
- Division of Diagnostic for Non-infectious Disease, National Institutes for Food and Drug Control, Beijing, China
| | - Nan Sun
- Division of Diagnostic for Non-infectious Disease, National Institutes for Food and Drug Control, Beijing, China
| | - Wenxin Zhang
- Division of Diagnostic for Non-infectious Disease, National Institutes for Food and Drug Control, Beijing, China
| | - Juanli Zhang
- Department of Invitro Diagnostic Reagents Testing, Henan Medical Equipment Inspection Institute, Zhengzhou, China
| | - Qingtao Song
- R&D Center, Amoy Diagnostics Co., Ltd, Xiamen, China
| | - Mufei Lin
- Oncology Business Unit, BGI Geonmics Co., Ltd, Shenzhen, China
| | - Wei Gao
- R&D Center, Geneplus-Beijing Institute, Beijing, China
| | - Qiaosong Zheng
- R&D Center, Genetron Health (Beijing) Co, Beijing, China
| | - Mipeng Han
- R&D Center, Berry Genomics Co., Ltd, Beijing, China
| | - Chenglong Na
- R&D Center, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Ren Xu
- R&D Center, Shanghai Yuanqi Bio-Pharmaceutical Co. Ltd, Shanghai, China
| | - Xiaoyan Chang
- Department of Pathology, Peking Union Medical College Hospital, Beijing, China
| | - Xuexi Yang
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Jie Huang
- Division of Diagnostic for Non-infectious Disease, National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
38
|
Du W, Sun L, Liu T, Zhu J, Zeng Y, Zhang Y, Wang X, Liu Z, Huang JA. The miR‑625‑3p/AXL axis induces non‑T790M acquired resistance to EGFR‑TKI via activation of the TGF‑β/Smad pathway and EMT in EGFR‑mutant non‑small cell lung cancer. Oncol Rep 2020; 44:185-195. [PMID: 32319651 PMCID: PMC7251657 DOI: 10.3892/or.2020.7579] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 03/20/2020] [Indexed: 02/06/2023] Open
Abstract
Gefitinib is currently the preferred treatment for non-small cell lung cancer (NSCLC) patients with epidermal growth factor receptor (EGFR)-activating mutation. However, some patients gradually develop acquired resistance after receiving treatment. In addition to secondary T790M mutation, the remaining mechanisms contributing to non-T790M mutations need to be explored. In the present study, NSCLC-derived HCC827 and PC-9 cells and the corresponding gefitinib-resistant cell lines (HCC827GR and PC9GR) were utilized. Next-generation DNA sequencing was performed on the HCC827GR and PC9GR cells. Under AXL receptor tyrosine kinase (AXL) knockdown or miR-625-3p overexpressing conditions, a cell growth inhibition assay was performed to evaluate gefitinib sensitivity. Wound healing and Transwell assays were used to examine the migratory and invasive abilities of the cells. Moreover, we also carried out western blot analysis to detect the altered downstream signaling pathway. Our study revealed markedly decreased miR-625-3p expression in the HCC827GR cell line, while its overexpression partly reversed gefitinib resistance. Integrated analysis based on Targetscan website showed that AXL can be potentially targeted by miR-625-3p and we further verified the hypothesis via dual-luciferase reporter assays. Mechanistic analysis revealed that TGF-β1-induced EMT may contribute to the miR-625-3p/AXL axis-mediated gefitinib resistance. Our data demonstrated that miR-625-3p contributes to the acquired resistance of gefitinib, which may provide novel insight to combat resistance to EGFR-TKIs.
Collapse
Affiliation(s)
- Wenwen Du
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Lin Sun
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Ting Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jianjie Zhu
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yuanyuan Zeng
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yang Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Xueting Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Zeyi Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jian-An Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
39
|
Decreasing Arl4c expression by inhibition of AKT signal in human lung adenocarcinoma cells. Life Sci 2020; 246:117428. [DOI: 10.1016/j.lfs.2020.117428] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 02/09/2020] [Accepted: 02/10/2020] [Indexed: 12/15/2022]
|
40
|
Network Pharmacology Study on the Pharmacological Mechanism of Cinobufotalin Injection against Lung Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:1246742. [PMID: 32148531 PMCID: PMC7048923 DOI: 10.1155/2020/1246742] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/17/2019] [Accepted: 01/07/2020] [Indexed: 02/06/2023]
Abstract
Cinobufotalin injection, extracted from the skin of Chinese giant salamander or black sable, has good clinical effect against lung cancer. However, owing to its complex composition, the pharmacological mechanism of cinobufotalin injection has not been fully clarified. This study aimed to explore the mechanism of action of cinobufotalin injection against lung cancer using network pharmacology and bioinformatics. Compounds of cinobufotalin injection were determined by literature retrieval, and potential therapeutic targets of cinobufotalin injection were screened from Swiss Target Prediction and STITCH databases. Lung-cancer-related genes were summarized from GeneCards, OMIM, and DrugBank databases. The pharmacological mechanism of cinobufotalin injection against lung cancer was determined by enrichment analysis of gene ontology and Kyoto Encyclopedia of Genes and Genomes, and protein-protein interaction network was constructed. We identified 23 compounds and 506 potential therapeutic targets of cinobufotalin injection, as well as 70 genes as potential therapeutic targets of cinobufotalin injection in lung cancer by molecular docking. The antilung cancer effect of cinobufotalin injection was shown to involve cell cycle, cell proliferation, antiangiogenesis effect, and immune inflammation pathways, such as PI3K-Akt, VEGF, and the Toll-like receptor signaling pathway. In network analysis, the hub targets of cinobufotalin injection against lung cancer were identified as VEGFA, EGFR, CCND1, CASP3, and AKT1. A network diagram of “drug-compounds-target-pathway” was constructed through network pharmacology to elucidate the pharmacological mechanism of the antilung cancer effect of cinobufotalin injection, which is conducive to guiding clinical medication.
Collapse
|
41
|
Yan M, Wang W, Zhou J, Chang M, Peng W, Zhang G, Li J, Li H, Bai C. Knockdown of PLAT enhances the anticancer effect of gefitinib in non-small cell lung cancer. J Thorac Dis 2020; 12:712-723. [PMID: 32274137 PMCID: PMC7139041 DOI: 10.21037/jtd.2019.12.106] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Tyrosine kinase inhibitors (TKIs), such as gefitinib, are widely used as standard treatments for non-small cell lung cancer (NSCLC) patients with epidermal growth factor receptor (EGFR) mutations. However, the subsequent inevitable drug resistance has become a major challenge in clinical treatment. The aim of this study was to investigate the role of tissue-type plasminogen activator (PLAT) in gefitinib resistance in NSCLC. Methods The function of PLAT was determined using gefitinib-resistant cells and a nude mouse model. The gene knockdown was achieved by Lentivirus based RNA silence technique. Expression of relevant genes and proteins, cell viability, proliferation, apoptosis, cell cycle, reactive oxygen species levels, mitochondrial membrane potential and differential gene expression was detected by RT-qPCR, western blot, cell counting kit-8 assay, EdU incorporation, flow cytometry, JC-1 dye assay and complementary DNA arrays. The effects of PLAT knockdown on tumorigenesis was analyzed in vivo. Results Gefitinib-resistant cells expressed higher levels of PLAT and that knockdown of PLAT in resistant cells restored gefitinib sensitivity. Tumor proliferation was limited in vivo following PLAT knockdown. Moreover, PLAT knockdown affected mitochondrial function, caused caspase activation and cell cycle arrest, and activated TNF-α signaling, leading to apoptosis of gefitinib-resistant PC9 cells. Conclusions Our results suggest that PLAT reduces apoptosis of NSCLC cells and knockdown of PLAT enhances anticancer effect of gefitinib by upregulating TNF-α signaling.
Collapse
Affiliation(s)
- Mengnan Yan
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wei Wang
- Department of Pathology, Affiliated Yantai Yuhuangding Hospital, Medical College of Qingdao University, Yantai 264000, China
| | - Jian Zhou
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Meijia Chang
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wenjun Peng
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ge Zhang
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jing Li
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Huayin Li
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chunxue Bai
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
42
|
Deng M, Liu B, Zhang Z, Chen Y, Wang Y, Wang X, Lv Q, Yang X, Hou K, Che X, Qu X, Liu Y, Zhang Y, Hu X. Loss of G-protein-signaling modulator 2 accelerates proliferation of lung adenocarcinoma via EGFR signaling pathway. Int J Biochem Cell Biol 2020; 122:105716. [PMID: 32058048 DOI: 10.1016/j.biocel.2020.105716] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/31/2020] [Accepted: 02/10/2020] [Indexed: 12/20/2022]
Abstract
G-protein-signaling modulator 2 (GPSM2) belongs to a protein family that regulates activation of G proteins and plays an important role in mitotic spindle orientation. However, the role of GPSM2 in lung adenocarcinoma (LUAD) is still unclear. In this study, it was found that GPSM2 correlates with clinicopathological features and patient's prognosis in LUAD. Knocking down GPSM2 promoted LUAD cell proliferation in vitro and in vivo. Mechanistically, it was demonstrated that GPSM2 knockdown accelerates cell proliferation via the EGFR pathway. These results confirmed that GPSM2 played an important role in LUAD. Moreover, GPSM2, as an independent prognostic factor, may serve as a potential drug target and prognostic biomarker in LUAD.
Collapse
Affiliation(s)
- Mingming Deng
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, China; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Bofang Liu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang, China
| | - Zhe Zhang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang 110001, China
| | - Yang Chen
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yizhe Wang
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, China
| | - Ximing Wang
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, China
| | - Qingjie Lv
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang 110001, China
| | - Xianghong Yang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang 110001, China
| | - Kezuo Hou
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiaofang Che
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Ye Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Xuejun Hu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
43
|
Anania MC, Di Marco T, Mazzoni M, Greco A. Targeting Non-Oncogene Addiction: Focus on Thyroid Cancer. Cancers (Basel) 2020; 12:cancers12010129. [PMID: 31947935 PMCID: PMC7017043 DOI: 10.3390/cancers12010129] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/21/2019] [Accepted: 12/24/2019] [Indexed: 12/12/2022] Open
Abstract
Thyroid carcinoma (TC) is the most common malignancy of endocrine organs with an increasing incidence in industrialized countries. The majority of TC are characterized by a good prognosis, even though cases with aggressive forms not cured by standard therapies are also present. Moreover, target therapies have led to low rates of partial response and prompted the emergence of resistance, indicating that new therapies are needed. In this review, we summarize current literature about the non-oncogene addiction (NOA) concept, which indicates that cancer cells, at variance with normal cells, rely on the activity of genes, usually not mutated or aberrantly expressed, essential for coping with the transformed phenotype. We highlight the potential of non-oncogenes as a point of intervention for cancer therapy in general, and present evidence for new putative non-oncogenes that are essential for TC survival and that may constitute attractive new therapeutic targets.
Collapse
|
44
|
Grossi F, Genova C, Crinò L, Delmonte A, Turci D, Signorelli D, Passaro A, Soto Parra H, Catino A, Landi L, Gelsomino F, Tiseo M, Puppo G, Roila F, Ricciardi S, Tonini G, Cognetti F, Toschi L, Tassinari D, Scoppola A, Giannarelli D, Cortesi E. Real-life results from the overall population and key subgroups within the Italian cohort of nivolumab expanded access program in non-squamous non–small cell lung cancer. Eur J Cancer 2019; 123:72-80. [DOI: 10.1016/j.ejca.2019.09.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 08/22/2019] [Accepted: 09/06/2019] [Indexed: 01/12/2023]
|
45
|
Zhu H, Wang XQ, Lu HY. The coexistence of ROS1-rearranged lung adenocarcinoma and pulmonary tuberculosis in a critical ill young patient. J Int Med Res 2019; 48:300060519883752. [PMID: 31779512 PMCID: PMC7607273 DOI: 10.1177/0300060519883752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The coexistence of lung cancer and pulmonary tuberculosis (PTB) is uncommon in young
patients. We report a case of 22-year-old man who presented with a one-month history of
chest pain, cough, slight haemoptysis and weight loss. Following two acid fast bacilli
positive sputum samples, a diagnosis of TB was concluded. However, his response to anti-TB
therapy was inadequate. A CT scan and further laboratory tests assisted the final
diagnosis as c-ros oncogene 1 (ROS1) rearranged lung adenocarcinoma and PTB. Despite
severe comorbidities, the patient achieved clinical remission following treatment with the
anti-cancer drug, crizotinib and anti- TB therapy. Clinicians should be aware that this
comorbidity can occur in all age groups and the clinical and radiological symptoms of the
two diseases are similar.
Collapse
Affiliation(s)
- Hui Zhu
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiao-Qin Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hai-Yan Lu
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
46
|
Aran V, Omerovic J. Current Approaches in NSCLC Targeting K-RAS and EGFR. Int J Mol Sci 2019; 20:E5701. [PMID: 31739412 PMCID: PMC6888213 DOI: 10.3390/ijms20225701] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/12/2019] [Accepted: 11/12/2019] [Indexed: 12/24/2022] Open
Abstract
The research and treatment of non-small cell lung cancer (NSCLC) have achieved some important advances in recent years. Nonetheless, the overall survival rates for NSCLC remain low, indicating the importance to effectively develop new therapies and improve current approaches. The understanding of the function of different biomarkers involved in NSCLC progression, survival and response to therapy are important for the development of early detection tools and treatment options. Epidermal growth factor receptor (EGFR) and Kirsten rat sarcoma viral oncogene homolog (K-RAS) are two of the main significant biomarkers for the management of NSCLC. Mutations in these genes were associated with development and response to therapies. For example, the use of small molecule tyrosine kinase (TK) inhibitors and immunotherapy has led to benefits in some, but not all patients with altered EGFR. In contrast, there is still no effective approved drug to act upon patients harbouring K-RAS mutations. In addition, K-RAS mutations have been associated with lack of activity of TK inhibitors. However, promising approaches aimed to inhibit mutant K-RAS are currently under study. Therefore, this review will discuss these approaches and also EGFR therapies, and hopefully, it will draw attention to the need of continued research in the field in order to improve the outcomes in NSCLC patients.
Collapse
Affiliation(s)
- Veronica Aran
- Research Division, National Institute of Traumatology and Orthopedics, Av. Brasil 500, 20940-070 Rio de Janeiro, Brazil
| | - Jasminka Omerovic
- School of Medicine, University of Split, Šoltanska 2, 21000 Split, Croatia;
| |
Collapse
|
47
|
Li F, Li X, Li Z, Ji W, Lu S, Xia W. βKlotho is identified as a target for theranostics in non-small cell lung cancer. Theranostics 2019; 9:7474-7489. [PMID: 31695781 PMCID: PMC6831461 DOI: 10.7150/thno.35582] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/11/2019] [Indexed: 12/11/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) remains a great challenge, calling for the identification of novel molecular targets with diagnostic/therapeutic value. Here, we sought to characterize the expression of βKlotho and its anti-tumor roles in NSCLC. Methods: The expression of βKlotho was examined in NSCLC cells and tissues by western blot, qRT-PCR and immunohistochemistry staining respectively. Biological roles of βKlotho were revealed by a series of functional in vitro and in vivo studies. Serum βKlotho concentrations of patients were measured using specific ELISA methods. Results: Serum βKlotho concentrations of NSCLC patients were significantly lower than the control group. Moreover, βKlotho expression was negatively associated with lymph node metastasis, overall survival and progression-free survival. Overexpression of βKlotho or exogenous βKlotho administration inhibited the proliferation and migration of NSCLC cells, accompanied by induction of apoptosis, G1 to S phase arrest, and inactivation of ERK1/2, AKT and STAT3 signaling. Furthermore, βKlotho overexpression inhibited NSCLC tumor growth in vivo. Conclusions: βKlotho serves as a novel target for theranostics in NSCLC, which has potential clinical applications in the future.
Collapse
Affiliation(s)
- Fan Li
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xiyao Li
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ziming Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wenxiang Ji
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Weiliang Xia
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
48
|
Sippel TR, Johnson AM, Li HY, Hanson D, Nguyen TT, Bullock BL, Poczobutt JM, Kwak JW, Kleczko EK, Weiser-Evans MC, Nemenoff RA. Activation of PPARγ in Myeloid Cells Promotes Progression of Epithelial Lung Tumors through TGFβ1. Mol Cancer Res 2019; 17:1748-1758. [PMID: 31088909 DOI: 10.1158/1541-7786.mcr-19-0236] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/11/2019] [Accepted: 05/10/2019] [Indexed: 12/13/2022]
Abstract
Lung cancer is a heterogeneous disease in which patient-specific treatments are desirable and the development of targeted therapies has been effective. Although mutations in KRAS are frequent in lung adenocarcinoma, there are currently no targeted agents against KRAS. Using a mouse lung adenocarcinoma cell line with a Kras mutation (CMT167), we previously showed that PPARγ activation in lung cancer cells inhibits cell growth in vitro yet promotes tumor progression when activated in myeloid cells of the tumor microenvironment. Here, we report that PPARγ activation in myeloid cells promotes the production of TGFβ1, which, in turn, acts on CMT167 cancer cells to increase migration and induce an epithelial-mesenchymal transition (EMT). Targeting TGFβ1 signaling in CMT167 cells prevented their growth and metastasis in vivo. Similarly, another mouse lung adenocarcinoma cell line with a Kras mutation, LLC, induced TGFβ1 in myeloid cells through PPARγ activation. However, LLC cells are more mesenchymal and did not undergo EMT in response to TGFβ1, nor did LLC require TGFβ1 signaling for metastasis in vivo. Converting CMT167 cells to a mesenchymal phenotype through overexpression of ZEB1 made them unresponsive to TGFβ1 receptor inhibition. The ability of TGFβ1 to induce EMT in lung tumors may represent a critical process in cancer progression. We propose that TGFβ receptor inhibition could provide an additional treatment option for KRAS-mutant epithelial lung tumors.Implications: This study suggests that TGFβ receptor inhibitors may be an effective therapy in a subset of KRAS-mutant patients with non-small cell lung cancer, which show an epithelial phenotype.
Collapse
Affiliation(s)
- Trisha R Sippel
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Amber M Johnson
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Howard Y Li
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado.,Department of Veterans Affairs Medical Center, Denver, Colorado
| | - Dwight Hanson
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Teresa T Nguyen
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Bonnie L Bullock
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Joanna M Poczobutt
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Jeff W Kwak
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Emily K Kleczko
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Mary C Weiser-Evans
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado.,Department of Veterans Affairs Medical Center, Denver, Colorado
| | - Raphael A Nemenoff
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado. .,Department of Veterans Affairs Medical Center, Denver, Colorado
| |
Collapse
|
49
|
Kleczko EK, Kwak JW, Schenk EL, Nemenoff RA. Targeting the Complement Pathway as a Therapeutic Strategy in Lung Cancer. Front Immunol 2019; 10:954. [PMID: 31134065 PMCID: PMC6522855 DOI: 10.3389/fimmu.2019.00954] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/15/2019] [Indexed: 12/20/2022] Open
Abstract
Lung cancer is the leading cause of cancer death in men and women. Lung adenocarcinoma (LUAD), represents approximately 40% of all lung cancer cases. Advances in recent years, such as the identification of oncogenes and the use of immunotherapies, have changed the treatment of LUAD. Yet survival rates still remain low. Additionally, there is still a gap in understanding the molecular and cellular interactions between cancer cells and the immune tumor microenvironment (TME). Defining how cancer cells with distinct oncogenic drivers interact with the TME and new strategies for enhancing anti-tumor immunity are greatly needed. The complement cascade, a central part of the innate immune system, plays an important role in regulation of adaptive immunity. Initially it was proposed that complement activation on the surface of cancer cells would inhibit cancer progression via membrane attack complex (MAC)-dependent killing. However, data from several groups have shown that complement activation promotes cancer progression, probably through the actions of anaphylatoxins (C3a and C5a) on the TME and engagement of immunoevasive pathways. While originally shown to be produced in the liver, recent studies show localized complement production in numerous cell types including immune cells and tumor cells. These results suggest that complement inhibitory drugs may represent a powerful new approach for treatment of NSCLC, and numerous new anti-complement drugs are in clinical development. However, the mechanisms by which complement is activated and affects tumor progression are not well understood. Furthermore, the role of local complement production vs. systemic activation has not been carefully examined. This review will focus on our current understanding of complement action in LUAD, and describe gaps in our knowledge critical for advancing complement therapy into the clinic.
Collapse
Affiliation(s)
- Emily K Kleczko
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jeff W Kwak
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Erin L Schenk
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Raphael A Nemenoff
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|