1
|
Wang X, Ding L, Sun S, Loo SK, Chen L, Li T, Wang MT, Pennathur A, Huang Y, Gao SJ. CASTOR1 : A Novel Tumor Suppressor Linking mTORC1 and KRAS Pathways in Tumorigenesis and Resistance to KRAS-Targeted Therapies in Non-Small Cell Lung Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.23.650349. [PMID: 40313924 PMCID: PMC12045348 DOI: 10.1101/2025.04.23.650349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Cytosolic arginine sensor for mTORC1 Subunit 1 (CASTOR1) functions as a key regulator of mechanistic target of rapamycin complex 1 (mTORC1) signaling. Despite its frequent dysregulation in cancers via mechanisms such as KSHV microRNA-mediated inhibition or AKT-driven phosphorylation and degradation, the impact of CASTOR1 loss on tumor initiation and progression remains poorly understood. Here, we identify CASTOR1 as a critical tumor suppressor in non-small cell lung cancer (NSCLC) by demonstrating that its genetic ablation amplifies tumorigenesis in a KRAS -driven genetically engineered mouse model (GEMM;LSL- KRAS G12D ). CASTOR1 deficiency markedly enhances lung tumor incidence, accelerates tumor progression, and increases proliferative indices in KRAS G12D -driven tumors ( KRAS G12D ; C1 KO ) compared to CASTOR1 wild type (WT) tumors ( KRAS G12D ; C1 WT ). Advanced-stage tumors exhibit elevated phosphorylated CASTOR1 (pCASTOR1) and reduced total CASTOR1 levels, suggesting active degradation during tumorigenesis. Mechanistically, CASTOR1 loss amplifies mTORC1 signaling, as evidenced by heightened phosphorylation of downstream effectors 4EBP1 and S6, while also augmenting AKT and ERK activation, uncovering a crosstalk between the PI3K/AKT/mTORC1 and KRAS/ERK pathways. Furthermore, CASTOR1 ablation induces genome instability, which may contribute to enhanced tumor incidence and progression. Importantly, CASTOR1 deficiency confers resistance to KRAS G12D -specific inhibitors, while over half of KRAS G12D ; C1 WT tumors also display resistance. Organoids derived from KRAS G12D ; C1 KO and KRAS G12D ; C1 WT tumors reveal a correlation between KRAS inhibitor resistance and hyperactivation of mTORC1, with mTORC1 and PI3K inhibitors sensitizing resistant tumors to KRAS G12D -targeted therapies. These findings position CASTOR1 as a novel tumor suppressor that modulates mTORC1 and KRAS signaling to constrain NSCLC progression. Our study further highlights the therapeutic potential of combining mTORC1 or ERK inhibitors with KRAS-targeted therapies for NSCLC characterized by hyperactive KRAS signaling and impaired CASTOR1 activity. Highlights CASTOR1 functions as a tumor suppressor in NSCLC by limiting KRAS -driven tumor initiation and progression. CASTOR1 is frequently lost or inactivated in wild-type tumors during tumor progression, contributing to advanced-stage malignancies.CASTOR1 deficiency amplifies mTORC1 signaling and enhances PI3K/AKT and KRAS/ERK crosstalk, driving tumorigenesis and resistance to KRAS-specific inhibitors. Combining mTORC1 or PI3K inhibitors with KRAS-targeted therapies effectively overcomes resistance in KRAS -driven NSCLC.
Collapse
|
2
|
Gargalionis AN, Papavassiliou KA, Papavassiliou AG. Implication of mTOR Signaling in NSCLC: Mechanisms and Therapeutic Perspectives. Cells 2023; 12:2014. [PMID: 37566093 PMCID: PMC10416991 DOI: 10.3390/cells12152014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/28/2023] [Accepted: 08/06/2023] [Indexed: 08/12/2023] Open
Abstract
Mechanistic target of the rapamycin (mTOR) signaling pathway represents a central cellular kinase that controls cell survival and metabolism. Increased mTOR activation, along with upregulation of respective upstream and downstream signaling components, have been established as oncogenic features in cancer cells in various tumor types. Nevertheless, mTOR pathway therapeutic targeting has been proven to be quite challenging in various clinical settings. Non-small cell lung cancer (NSCLC) is a frequent type of solid tumor in both genders, where aberrant regulation of the mTOR pathway contributes to the development of oncogenesis, apoptosis resistance, angiogenesis, cancer progression, and metastasis. In this context, the outcome of mTOR pathway targeting in clinical trials still demonstrates unsatisfactory results. Herewith, we discuss recent findings regarding the mechanisms and therapeutic targeting of mTOR signaling networks in NSCLC, as well as future perspectives for the efficient application of treatments against mTOR and related protein molecules.
Collapse
Affiliation(s)
- Antonios N. Gargalionis
- Department of Biopathology, ‘Eginition’ Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Kostas A. Papavassiliou
- First University Department of Respiratory Medicine, ‘Sotiria’ Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
3
|
Kiełbowski K, Ptaszyński K, Wójcik J, Wojtyś ME. The role of selected non-coding RNAs in the biology of non-small cell lung cancer. Adv Med Sci 2023; 68:121-137. [PMID: 36933328 DOI: 10.1016/j.advms.2023.02.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 11/26/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023]
Abstract
Lung cancer is the second most frequently diagnosed cancer worldwide and a leading cause of cancer-related deaths. Non-small cell lung carcinoma (NSCLC) represents 85% of all cases. Accumulating evidence highlights the outstanding role of non-coding RNA (ncRNA) in regulating the tumorigenesis process by modulating crucial signaling pathways. Micro RNA (miRNA), long non-coding RNA (lncRNA) and circular RNA (circRNA) are either up- or downregulated in lung cancer patients and can promote or suppress the progression of the disease. These molecules interact with messenger RNA (mRNA) and with each other to regulate gene expression and stimulate proto-oncogenes or silence tumor suppressors. NcRNAs provide a new strategy to diagnose or treat lung cancer patients and multiple molecules have already been identified as potential biomarkers or therapeutic targets. The aim of this review is to summarize the current evidence on the roles of miRNA, lncRNA and circRNA in NSCLC biology and present their clinical potential.
Collapse
Affiliation(s)
- Kajetan Kiełbowski
- Department of Thoracic Surgery and Transplantation, Pomeranian Medical University, Szczecin, Poland
| | - Konrad Ptaszyński
- Department of Pathology, Faculty of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Poland
| | - Janusz Wójcik
- Department of Thoracic Surgery and Transplantation, Pomeranian Medical University, Szczecin, Poland
| | - Małgorzata Edyta Wojtyś
- Department of Thoracic Surgery and Transplantation, Pomeranian Medical University, Szczecin, Poland.
| |
Collapse
|
4
|
Qiu W, Ren M, Wang C, Fu Y, Liu Y. The clinicopathological and prognostic significance of mTOR and p-mTOR expression in patients with non-small cell lung cancer: A meta-analysis. Medicine (Baltimore) 2022; 101:e32340. [PMID: 36595789 PMCID: PMC9794261 DOI: 10.1097/md.0000000000032340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/30/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The mammalian target of rapamycin (mTOR) has a crucial role in carcinogenesis, angiogenesis, cellular proliferation, and metastasis; however, its significance in non-small cell lung cancer (NSCLC) remains contentious. Consequently, this study aims to assess the clinicopathological and prognostic importance of mTOR/p-mTOR expression in NSCLC. METHODS Literature retrieval was undertaken by searching English databases PubMed, EMBASE, Web of Science, and Cochrane Library as well as Chinese databases CNKI, Wan Fang, and VIP for full-text publications that satisfied our eligibility criteria up to November 2021. STATA 12.0 was used to conduct statistical analysis (STATA Corporation, College Station, TX). RESULTS This meta-analysis includes a total of 4683 patients from 28 primary publications. mTOR/p-mTOR expression was associated with sex (OR = 0.608, 95% CI: 0.442-0.836), lymph node metastasis (OR = 2.084, 95% CI: 1.437-3.182), and CEA (OR = 1.584, 95% CI: 1.135-2.209), but not with age, histological type, depth of tumor invasion, distant metastasis, TNM stage, differentiation degree, tumor size, or smoking. In addition, the expression of mTOR/p-mTOR is related to shorter overall survival in NSCLC patients (HR = 1.415, 95% CI: 1.051-1.905). CONCLUSION Positive mTOR/p-mTOR expression was substantially correlated with unfavorable conditions on the sex, lymph node metastases, and CEA levels. mTOR/p-mTOR may indicate a bad prognosis for NSCLC. The current findings must be confirmed and changed by other high-quality research employing a multivariate analysis on bigger sample size.
Collapse
Affiliation(s)
- Weiwei Qiu
- Department of Laboratory Medicine, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
- Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Meiying Ren
- Department of Laboratory Medicine, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Cuifeng Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Yuhua Fu
- Department of Laboratory Medicine, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Yan Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| |
Collapse
|
5
|
Wang C, Aikemu B, Shao Y, Zhang S, Yang G, Hong H, Huang L, Jia H, Yang X, Zheng M, Sun J, Li J. Genomic signature of MTOR could be an immunogenicity marker in human colorectal cancer. BMC Cancer 2022; 22:818. [PMID: 35883111 PMCID: PMC9327395 DOI: 10.1186/s12885-022-09901-w] [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: 01/12/2022] [Accepted: 07/13/2022] [Indexed: 12/24/2022] Open
Abstract
Background The mTOR signaling pathway plays an important role in cancer. As a master regulator, the status of MTOR affects pathway activity and the efficacy of mTOR inhibitor therapy. However, little research has been performed to explore MTOR in colorectal cancer (CRC). Methods In this study, gene expression and clinical data were analyzed using The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases. Signaling pathways related to MTOR in CRC were identified by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA). Somatic mutation data were downloaded from TCGA and analyzed using the maftools R package. Tumor Immune Estimation Resource (TIMER) and CIBERSORT were used to analyze correlations between MTOR and tumor-infiltrating immune cells (TIICs). Finally, we detected MTOR mutations in a CRC cohort from our database using whole-exome sequencing. Results We found that MTOR was overexpressed in Asian CRC patients and associated with a poor prognosis. Enrichment analysis showed that MTOR was involved in metabolism, cell adhesion, and translation pathways in CRC. High MTOR expression was correlated with high tumor mutation burden (TMB) and several TIICs. Finally, we found that the mTOR signaling pathway was activated in CRC lines characterized by microsatellite instability (MSI), and the frequency of MTOR mutations was higher in MSI-high (MSI-H) patients than in microsatellite stable (MSS) patients. Conclusions MTOR may represent a comprehensive indicator of prognosis and immunological status in CRC. The genomic signatures of MTOR may provide guidance for exploring the role of mTOR inhibitors in CRC. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09901-w.
Collapse
Affiliation(s)
- Chenxing Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Batuer Aikemu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yanfei Shao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Sen Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Guang Yang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hiju Hong
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ling Huang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hongtao Jia
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiao Yang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Minhua Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jing Sun
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Jianwen Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
6
|
The sodium/myo-inositol co-transporter SLC5A3 promotes non-small cell lung cancer cell growth. Cell Death Dis 2022; 13:569. [PMID: 35760803 PMCID: PMC9237060 DOI: 10.1038/s41419-022-05017-y] [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: 01/10/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 01/21/2023]
Abstract
Identification of novel molecular signaling targets for non-small cell lung cancer (NSCLC) is important. The present study examined expression, functions and possible underlying mechanisms of the sodium/myo-inositol co-transporter SLC5A3 in NSCLC. The Cancer Genome Atlas (TCGA) database and local NSCLC tissue results demonstrated that SLC5A3 expression in NSCLC tissues (including patient-derived primary NSCLC cells) was significantly higher than that in normal lung tissues and lung epithelial cells. In primary NSCLC cells and immortalized lines, SLC5A3 depletion, using small hairpin RNA (shRNA) and CRSIRP/Cas9 methods, robustly impeded cell proliferation and migration, simultaneously provoking cell cycle arrest and apoptosis. Conversely, ectopic overexpression of SLC5A3 further enhanced proliferation and migration in primary NSCLC cells. The intracellular myo-inositol contents and Akt-mTOR activation were largely inhibited by SLC5A3 silencing or knockout (KO), but were augmented following SLC5A3 overexpression in primary NSCLC cells. Significantly, SLC5A3 KO-induced anti-NSCLC cell activity was largely ameliorated by exogenously adding myo-inositol or by a constitutively-active Akt construct. By employing the patient-derived xenograft (PDX) model, we found that the growth of subcutaneous NSCLC xenografts in nude mice was largely inhibited by intratumoral injection SLC5A3 shRNA adeno-associated virus (AAV). SLC5A3 silencing, myo-inositol depletion, Akt-mTOR inactivation and apoptosis induction were detected in SLC5A3 shRNA virus-injected NSCLC xenograft tissues. Together, elevated SLC5A3 promotes NSCLC cell growth possibly by maintaining myo-inositol contents and promoting Akt-mTOR activation.
Collapse
|
7
|
An mTOR and DNA-PK dual inhibitor CC-115 hinders non-small cell lung cancer cell growth. Cell Death Dis 2022; 8:293. [PMID: 35717530 PMCID: PMC9206683 DOI: 10.1038/s41420-022-01082-6] [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: 03/17/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022]
Abstract
Molecularly-targeted agents are still urgently needed for better non-small cell lung cancer (NSCLC) therapy. CC-115 is a potent DNA-dependent protein kinase (DNA-PK) and mammalian target of rapamycin (mTOR) dual blocker. We evaluated its activity in different human NSCLC cells. In various primary human NSCLC cells and A549 cells, CC-115 potently inhibited viability, cell proliferation, cell cycle progression, and hindered cell migration/invasion. Apoptosis was provoked in CC-115-stimulated NSCLC cells. The dual inhibitor, however, was unable to induce significant cytotoxic and pro-apoptotic activity in the lung epithelial cells. In primary NSCLC cells, CC-115 blocked activation of mTORC1/2 and DNA-PK. Yet, CC-115-induced primary NSCLC cell death was more potent than combined inhibition of DNA-PK plus mTOR. Further studies found that CC-115 provoked robust oxidative injury in primary NSCLC cells, which appeared independent of mTOR-DNA-PK dual blockage. In vivo studies showed that CC-115 oral administration in nude mice remarkably suppressed primary NSCLC cell xenograft growth. In CC-115-treated NSCLC xenograft tissues, mTOR-DNA-PK dual inhibition and oxidative injury were detected. Together, CC-115 potently inhibits NSCLC cell growth.
Collapse
|
8
|
Scopetti D, Piobbico D, Brunacci C, Pieroni S, Bellezza G, Castelli M, Ludovini V, Tofanetti FR, Cagini L, Sidoni A, Puxeddu E, Della-Fazia MA, Servillo G. INSL4 as prognostic marker for proliferation and invasiveness in Non-Small-Cell Lung Cancer. J Cancer 2021; 12:3781-3795. [PMID: 34093787 PMCID: PMC8176261 DOI: 10.7150/jca.51332] [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: 01/31/2021] [Accepted: 02/25/2021] [Indexed: 12/24/2022] Open
Abstract
Non-small-cell-lung cancer accounts for 80-85% of all forms of lung cancer as leading cause of cancer-related death in human. Despite remarkable advances in the diagnosis and therapy of lung cancer, no significant improvements have thus far been achieved in terms of patients' prognosis. Here, we investigated the role of INSL4 - a member of the relaxin-family - in NSCLC. We overexpressed INSL4 in NSCLC cells to analyse in vitro the growth rate and the tumourigenic features. We investigated the signalling pathways engaged in INSL4 overexpressing cells and the tumour growth ability by studying the tumour development in a patient derived tumour xenograft mouse model. We found an INSL4 cell growth promoting effect in vitro in H1299 cells and in vivo in NOD/SCID mice. Surprisingly, in NSCLC-A549 cells, INSL4 overexpression has not similar effect, despite huge basal INSL4-mRNA expression respect to H1299. The INSL4-mRNA analysis of eight different NSCLC-derived cell lines, revealed highly difference in the INSL4-mRNA amount. Transfection of NSCLC lines with INSL4-Myc showed huge level of INSL4-mRNA with a very low amount of protein expressed. Notably, similar discrepancy has been observed in NSCLC patients. However, in a cohort of NSCLC patients analysing a database, we found a significant inverse correlation between INSL4 expression and Overall Survival. By combining the in vitro and in vivo results, suggest that in patients whose NSCLC adenocarcinoma spontaneously expressed high levels of INSL4 post-transcriptional modifications affecting INSL4 do not allow to assess precision therapy in selected patients without consider protein INSL4 amount.
Collapse
Affiliation(s)
- Damiano Scopetti
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| | - Danilo Piobbico
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| | - Cinzia Brunacci
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| | - Stefania Pieroni
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| | - Guido Bellezza
- Department of Medicine and Surgery, Section of Anatomic Pathology and Histology, University of Perugia, Perugia- Italy
| | - Marilena Castelli
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| | - Vienna Ludovini
- Medical Oncology, S. Maria Della Misericordia Hospital, Perugia, Italy
| | | | - Lucio Cagini
- Department of Medicine and Surgery, Section of internal medicine, angiology and atherosclerosis diseases
| | - Angelo Sidoni
- Department of Medicine and Surgery, Section of Anatomic Pathology and Histology, University of Perugia, Perugia- Italy
| | - Efisio Puxeddu
- Department of Medicine and Surgery, Section of internal medicine and endocrine and metabolic sciences
| | - Maria Agnese Della-Fazia
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| | - Giuseppe Servillo
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| |
Collapse
|
9
|
PM014 attenuates radiation-induced pulmonary fibrosis via regulating NF-kB and TGF-b1/NOX4 pathways. Sci Rep 2020; 10:16112. [PMID: 32999298 PMCID: PMC7527517 DOI: 10.1038/s41598-020-72629-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 09/04/2020] [Indexed: 12/31/2022] Open
Abstract
Radiation therapy is the mainstay in the treatment of lung cancer, and lung fibrosis is a radiotherapy-related major side effect that can seriously reduce patient’s quality of life. Nevertheless, effective strategies for protecting against radiation therapy-induced fibrosis have not been developed. Hence, we investigated the radioprotective effects and the underlying mechanism of the standardized herbal extract PM014 on radiation-induced lung fibrosis. Ablative radiation dose of 75 Gy was focally delivered to the left lung of mice. We evaluated the effects of PM014 on radiation-induced lung fibrosis in vivo and in an in vitro model. Lung volume and functional changes were evaluated using the micro-CT and flexiVent system. Fibrosis-related molecules were evaluated by immunohistochemistry, western blot, and real-time PCR. A orthotopic lung tumour mouse model was established using LLC1 cells. Irradiated mice treated with PM014 showed a significant improvement in collagen deposition, normal lung volume, and functional lung parameters, and these therapeutic effects were better than those of amifostine. PM104 attenuated radiation-induced increases in NF-κB activity and inhibited radiation-induced p65 translocation, ROS production, DNA damage, and epithelial-mesenchymal transition. PM104 effectively alleviated fibrosis in an irradiated orthotopic mouse lung tumour model while not attenuating the efficacy of the radiation therapy by reduction of the tumour. Standardized herbal extract PM014 may be a potential therapeutic agent that is able to increase the efficacy of radiotherapy by alleviating radiation-induced lung fibrosis.
Collapse
|
10
|
Crees ZD, Shearrow C, Lin L, Girard J, Arasi K, Bhoraskar A, Berei J, Eckburg A, Anderson AD, Garcia C, Munger A, Palani S, Smith TJ, Sreenivassappa SB, Vitali C, David O, Puri N. EGFR/c-Met and mTOR signaling are predictors of survival in non-small cell lung cancer. Ther Adv Med Oncol 2020; 12:1758835920953731. [PMID: 32973931 PMCID: PMC7493230 DOI: 10.1177/1758835920953731] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 08/06/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND EGFR/c-Met activation/amplification and co-expression, mTOR upregulation/activation, and Akt/Wnt signaling upregulation have been individually associated with more aggressive disease and characterized as potential prognostic markers for lung cancer patients. METHODS Tumors obtained from 109 participants with stage I-IV non-small cell lung cancer (NSCLC) were studied for EGFR/c-Met co-localization as well as for total and active forms of EGFR, c-Met, mTOR, S6K, beta-catenin, and Axin2. Slides were graded by two independent blinded pathologists using a validated scoring system. Protein expression profile correlations were assessed using Pearson correlation and Spearman's rho. Prognosis was assessed using Kaplan-Meier analysis. RESULTS Protein expression profile analysis revealed significant correlations between EGFR/p-EGFR (p = 0.0412) and p-mTOR/S6K (p = 0.0044). Co-localization of p-EGFR/p-c-Met was associated with increased p-mTOR (p = 0.0006), S6K (p = 0.0018), and p-S6K (p < 0.0001) expression. In contrast, active beta-catenin was not positively correlated with EGFR/c-Met nor any activated proteins. Axin2, a negative regulator of the Wnt pathway, was correlated with EGFR, p-EGFR, p-mTOR, p-S6K, EGFR/c-Met co-localization, and p-EGFR/p-c-Met co-localization (all p-values <0.03). Kaplan-Meier analysis revealed shorter median survival in participants with high expression of Axin2, total beta-catenin, total/p-S6K, total/p-mTOR, EGFR, and EGFR/c-Met co-localization compared with low expression. After controlling for stage of disease at diagnosis, subjects with late-stage disease demonstrated shorter median survival when exhibiting high co-expression of EGFR/c-Met (8.1 month versus 22.3 month, p = 0.050), mTOR (6.7 month versus 22.3 month, p = 0.002), and p-mTOR (8.1 month versus 25.4 month, p = 0.004) compared with low levels. CONCLUSIONS These findings suggest that increased EGFR/c-Met signaling is correlated with upregulated mTOR/S6K signaling, which may in turn be associated with shorter median survival in late-stage NSCLC.
Collapse
Affiliation(s)
- Zachary D Crees
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, IL, USA
| | - Caleb Shearrow
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, IL, USA
| | - Leo Lin
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, IL, USA
| | - Jennifer Girard
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, IL, USA
| | - Kavin Arasi
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, IL, USA
| | - Aayush Bhoraskar
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, IL, USA
| | - Joseph Berei
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, IL, USA
| | - Adam Eckburg
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, IL, USA
| | - Austin D. Anderson
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, IL, USA
| | - Christian Garcia
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, IL, USA
| | - Ariana Munger
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, IL, USA
| | - Sunil Palani
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, IL, USA
| | - Thomas J Smith
- College of Education, Northern Illinois University, Dekalb, IL, USA
| | | | - Connie Vitali
- Department of Pathology, University of Illinois College of Medicine at Rockford IL, USA
| | - Odile David
- Department of Pathology, University of Illinois College of Medicine at Chicago, IL, USA
| | - Neelu Puri
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, 1601 Parkview Avenue, Room Number E-632, Rockford, IL 61107, USA
| |
Collapse
|
11
|
Tan AC. Targeting the PI3K/Akt/mTOR pathway in non-small cell lung cancer (NSCLC). Thorac Cancer 2020; 11:511-518. [PMID: 31989769 PMCID: PMC7049515 DOI: 10.1111/1759-7714.13328] [Citation(s) in RCA: 328] [Impact Index Per Article: 65.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 01/07/2020] [Indexed: 12/24/2022] Open
Abstract
The traditional classification of lung cancer into small cell lung cancer and non-small cell lung cancer (NSCLC) has been transformed with the increased understanding of the molecular alterations and genomic biomarkers that drive the development of lung cancer. Increased activation of the phosphatidylinositol 3-kinase (PI3K)/Akt/mechanistic target of rapamycin (mTOR) pathway leads to numerous hallmarks of cancer and this pathway represents an attractive target for novel anticancer therapies. In NSCLC, the PI3K/Akt/mTOR pathway has been heavily implicated in both tumorigenesis and the progression of disease. A number of specific inhibitors of PI3K, Akt and mTOR are currently under development and in various stages of preclinical investigation and in early phase clinical trials for NSCLC. Early evidence has yielded disappointing results. Clinical trials, however, have been performed on predominantly molecularly unselected populations, and patient enrichment strategies using high-precision predictive biomarkers in future trials will increase the likelihood of success. A greater understanding of the underlying molecular biology including epigenetic alterations is also crucial to allow for the detection of appropriate biomarkers and guide combination approaches.
Collapse
Affiliation(s)
- Aaron C. Tan
- Division of Medical OncologyNational Cancer Centre SingaporeSingapore
| |
Collapse
|
12
|
Galetta D, Pizzutilo P, Longo V. BRMS1 expression in resected lung adenocarcinoma. Transl Lung Cancer Res 2018; 7:S364-S366. [PMID: 30705857 DOI: 10.21037/tlcr.2018.09.20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Domenico Galetta
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Pamela Pizzutilo
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Vito Longo
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| |
Collapse
|
13
|
Dubash SR, Merchant S, Heinzmann K, Mauri F, Lavdas I, Inglese M, Kozlowski K, Rama N, Masrour N, Steel JF, Thornton A, Lim AK, Lewanski C, Cleator S, Coombes RC, Kenny L, Aboagye EO. Clinical translation of [ 18F]ICMT-11 for measuring chemotherapy-induced caspase 3/7 activation in breast and lung cancer. Eur J Nucl Med Mol Imaging 2018; 45:2285-2299. [PMID: 30259091 PMCID: PMC6208806 DOI: 10.1007/s00259-018-4098-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 07/17/2018] [Indexed: 01/17/2023]
Abstract
BACKGROUND Effective anticancer therapy is thought to involve induction of tumour cell death through apoptosis and/or necrosis. [18F]ICMT-11, an isatin sulfonamide caspase-3/7-specific radiotracer, has been developed for PET imaging and shown to have favourable dosimetry, safety, and biodistribution. We report the translation of [18F]ICMT-11 PET to measure chemotherapy-induced caspase-3/7 activation in breast and lung cancer patients receiving first-line therapy. RESULTS Breast tumour SUVmax of [18F]ICMT-11 was low at baseline and unchanged following therapy. Measurement of M30/M60 cytokeratin-18 cleavage products showed that therapy was predominantly not apoptosis in nature. While increases in caspase-3 staining on breast histology were seen, post-treatment caspase-3 positivity values were only approximately 1%; this low level of caspase-3 could have limited sensitive detection by [18F]ICMT-11-PET. Fourteen out of 15 breast cancer patients responded to first-line chemotherapy (complete or partial response); one patient had stable disease. Four patients showed increases in regions of high tumour [18F]ICMT-11 intensity on voxel-wise analysis of tumour data (classed as PADS); response was not exclusive to patients with this phenotype. In patients with lung cancer, multi-parametric [18F]ICMT-11 PET and MRI (diffusion-weighted- and dynamic contrast enhanced-MRI) showed that PET changes were concordant with cell death in the absence of significant perfusion changes. CONCLUSION This study highlights the potential use of [18F]ICMT-11 PET as a promising candidate for non-invasive imaging of caspase3/7 activation, and the difficulties encountered in assessing early-treatment responses. We summarize that tumour response could occur in the absence of predominant chemotherapy-induced caspase-3/7 activation measured non-invasively across entire tumour lesions in patients with breast and lung cancer.
Collapse
Affiliation(s)
- S R Dubash
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
| | - S Merchant
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
| | - K Heinzmann
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
| | - F Mauri
- Department of Radiology, Imperial College Healthcare NHS Trust, London, UK
| | - I Lavdas
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
| | - M Inglese
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
- Department of Computer, Control and Management Engineering Antonio Ruberti, University of Rome, La Sapienza, Italy
| | - K Kozlowski
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
| | - N Rama
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
| | - N Masrour
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
| | - J F Steel
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
| | - A Thornton
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK
| | - A K Lim
- Department of Radiology, Imperial College Healthcare NHS Trust, London, UK
| | - C Lewanski
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - S Cleator
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - R C Coombes
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - Laura Kenny
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK.
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK.
| | - Eric O Aboagye
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Rd, London, W120NN, UK.
| |
Collapse
|
14
|
Bellezza G, Vannucci J, Bianconi F, Metro G, Del Sordo R, Andolfi M, Ferri I, Siccu P, Ludovini V, Puma F, Sidoni A, Cagini L. Prognostic implication of aquaporin 1 overexpression in resected lung adenocarcinoma. Interact Cardiovasc Thorac Surg 2018; 25:856-861. [PMID: 29106595 DOI: 10.1093/icvts/ivx202] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 05/24/2017] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES Aquaporins (AQPs) are a group of transmembrane water-selective channel proteins thought to play a role in the regulation of water permeability for plasma membranes. Indeed, high AQP levels have been suggested to promote the progression, invasion and metastasis of tumours. Specifically, AQP1 and AQP5 overexpression in lung adenocarcinoma (AC) have been suggested to be involved in molecular mechanisms in lung cancer. The aim of this retrospective cohort single-centre study was to assess both the levels of expression and therein the prognostic significance, regarding outcome of AQP1 and AQP5 in resected AC patients. METHODS Patients with histological diagnoses of lung AC submitted to pulmonary resection were included in this cohort study. Tissue microarrays containing cores from 185 ACs were prepared. AQP1 and AQP5 expressions were assessed by immunohistochemistry. Results were scored as either low (Score 0-2) or high (Score 3-9). Clinical data, pathological tumour-node-metastasis staging and follow-up were recorded. Multivariate Cox survival analysis and Fisher's t-test were performed. RESULTS AQP1 overexpression was detected in 85 (46%) patients, while AQP5 overexpression was observed in 45 (24%) patients. AQP1 did not result being significantly correlated with clinical and pathological parameters, while AQP5 resulted more expressed in AC with mucinous and papillary predominant patterns. Patients with AQP1 overexpression had shorter disease-free survival (P = 0.001) compared with patients without AQP1 overexpression. Multivariate analysis confirmed that AQP1 overexpression was significantly associated with shorter disease-free survival (P = 0.001). CONCLUSIONS Our results evidenced that AQP1 overexpression resulted in a shorter disease-free survival in lung AC patients. Being so, AQP1 overexpression might be an important prognostic marker in lung AC.
Collapse
Affiliation(s)
- Guido Bellezza
- Section of Anatomic Pathology and Histology, Department of Experimental Medicine, University of Perugia Medical School, Perugia, Italy
| | - Jacopo Vannucci
- Department of Thoracic Surgery, University of Perugia Medical School, Perugia, Italy
| | - Fortunato Bianconi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Giulio Metro
- Division of Medical Oncology, S. Maria della Misericordia Hospital, Perugia, Italy
| | - Rachele Del Sordo
- Section of Anatomic Pathology and Histology, Department of Experimental Medicine, University of Perugia Medical School, Perugia, Italy
| | - Marco Andolfi
- Department of Thoracic Surgery, University of Perugia Medical School, Perugia, Italy
| | - Ivana Ferri
- Section of Anatomic Pathology and Histology, Department of Experimental Medicine, University of Perugia Medical School, Perugia, Italy
| | - Paola Siccu
- Section of Anatomic Pathology and Histology, Department of Experimental Medicine, University of Perugia Medical School, Perugia, Italy
| | - Vienna Ludovini
- Division of Medical Oncology, S. Maria della Misericordia Hospital, Perugia, Italy
| | - Francesco Puma
- Department of Thoracic Surgery, University of Perugia Medical School, Perugia, Italy
| | - Angelo Sidoni
- Section of Anatomic Pathology and Histology, Department of Experimental Medicine, University of Perugia Medical School, Perugia, Italy
| | - Lucio Cagini
- Department of Thoracic Surgery, University of Perugia Medical School, Perugia, Italy
| |
Collapse
|
15
|
Rupniewska E, Roy R, Mauri FA, Liu X, Kaliszczak M, Bellezza G, Cagini L, Barbareschi M, Ferrero S, Tommasi AM, Aboagye E, Seckl MJ, Pardo OE. Targeting autophagy sensitises lung cancer cells to Src family kinase inhibitors. Oncotarget 2018; 9:27346-27362. [PMID: 29937990 PMCID: PMC6007948 DOI: 10.18632/oncotarget.25213] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 04/04/2018] [Indexed: 11/25/2022] Open
Abstract
Lung cancer is the main cancer killer in both men and women, mostly due to the rapid development of drug resistant metastatic disease. Here, we evaluate the potential involvement of SRC family kinases (SFK) in lung cancer biology and assess the possible benefits of their inhibition as a therapeutic approach. We demonstrated that various SRC family members, including LYN and LCK, normally expressed solely in hematopoietic cells and neural tissues, are overexpressed and activated in a panel of SCLC and NSCLC cell lines. This was clinically relevant as LYN and FYN are also overexpressed in lung cancer clinical specimens. Moreover, LYN overexpression correlated with decreased patient survival on univariate and multivariate analysis. Dasatinib (BMS-354825), a SRC/ABL inhibitor, effectively blocked SFK activation at nanomolar concentrations which correlated with a significant decrease in cell numbers of multiple lung cancer cell lines. This effect was matched by a decrease in DNA synthesis, but only moderate induction of apoptosis. Indeed, dasatinib as well as PP2, another SFK inhibitor, strongly induced autophagy that likely prevented apoptosis. However, inhibition of this autophagic response induced robust apoptosis and sensitised lung cancer cells to dasatinib in vitro and in vivo. Our results provide an explanation for why dasatinib failed in NSCLC clinical trials. Furthermore, our data suggest that combining SFK inhibitors with autophagy inhibitors could provide a novel therapeutic approach in this disease.
Collapse
Affiliation(s)
- Ewa Rupniewska
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Rajat Roy
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Francesco A Mauri
- Department of Histopathology and Imperial College London, London, United Kingdom
| | - Xinxue Liu
- Statistical Advisory Service, Imperial College London, London, United Kingdom
| | - Maciej Kaliszczak
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Guido Bellezza
- Institute of Pathology, Division of Cancer Research, Perugia Medical School, University of Perugia, Perugia, Italy
| | - Lucio Cagini
- Department of Thoracic Surgery, Division of Cancer Research, Perugia Medical School, University of Perugia, Perugia, Italy
| | - Mattia Barbareschi
- Unit of Surgical Pathology, Laboratory of Molecular Pathology S. Chiara Hospital, Trento, Italy
| | - Stefano Ferrero
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Anna M Tommasi
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Eric Aboagye
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Michael J Seckl
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Olivier E Pardo
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| |
Collapse
|
16
|
Chandrasekaran B, Tyagi A, Sharma AK, Cai L, Ankem M, Damodaran C. Molecular insights: Suppression of EGFR and AKT activation by a small molecule in non-small cell lung cancer. Genes Cancer 2017; 8:713-724. [PMID: 29234489 PMCID: PMC5724805 DOI: 10.18632/genesandcancer.154] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 10/17/2017] [Indexed: 01/08/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) activation events and the mammalian target of rampamycin (mTOR) are considered important therapeutic targets in alleviating cancer conditions. The current treatment paradigm has shifted to personalized treatment strategies with tyrosine kinase inhibitors (TKIs) or anaplastic lymphoma kinase (ALK) inhibitors, due to low survival rates in non-small cell lung cancer (NSCLC) in terms of the prevailing platinum-based therapy. In the present study, we examined the anticancer potential of Verrucarin J (VJ), a small molecule, in NSCLC cell lines (H460 and A549). The small molecule significantly inhibited cell growth, proliferation, colony forming ability, and induced apoptosis in both lung cancer cell lines. The inhibitory effects on EGFR (pEGFR -tyr1173) and AKT (pAKT Serine473) signaling, downregulates downstream pro-survival signaling (mTOR and NF-κB) in cancer cell lines. In addition, VJ abrogated invasive and migratory potential of A549 and H460 cells. We also observed a downregulation of mesenchymal markers such as N-cadherin, Slug, β-catenin, and vimentin expression in both cell lines. Our results suggest that VJ inhibited cancer cell growth and could be a potent molecule to inhibit EGFR and AKT signaling in NSCLC.
Collapse
Affiliation(s)
| | - Ashish Tyagi
- Department of Urology, University of Louisville, Louisville, KY, USA
| | - Arun K. Sharma
- Department of Pharmacology, Penn State University, Hershey, PA, USA
| | - Lu Cai
- Pediatrics Research Institute, The Department of Pediatrics of the University of Louisville, Louisville, USA
| | - Murali Ankem
- Department of Urology, University of Louisville, Louisville, KY, USA
| | - Chendil Damodaran
- Department of Urology, University of Louisville, Louisville, KY, USA
| |
Collapse
|
17
|
Standardized Herbal Formula PM014 Inhibits Radiation-Induced Pulmonary Inflammation in Mice. Sci Rep 2017; 7:45001. [PMID: 28322297 PMCID: PMC5359558 DOI: 10.1038/srep45001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 02/17/2017] [Indexed: 12/27/2022] Open
Abstract
Radiation therapy is widely used for thoracic cancers. However, it occasionally causes radiation-induced lung injuries, including pneumonitis and fibrosis. Chung-Sang-Bo-Ha-Tang (CSBHT) has been traditionally used to treat chronic pulmonary disease in Korea. PM014, a modified herbal formula derived from CSBHT, contains medicinal herbs of seven species. In our previous studies, PM014 exhibited anti-inflammatory effects in a chronic obstructive pulmonary disease model. In this study, we have evaluated the effects of PM014 on radiation-induced lung inflammation. Mice in the treatment group were orally administered PM014 six times for 2 weeks. Effects of PM014 on radiation pneumonitis were evaluated based on histological findings and differential cell count in bronchoalveolar lavage fluid. PM014 treatment significantly inhibited immune cell recruitment and collagen deposition in lung tissue. Normal lung volume, evaluated by radiological analysis, in PM014-treated mice was higher compared to that in irradiated control mice. PM014-treated mice exhibited significant changes in inspiratory capacity, compliance and tissue damping and elastance. Additionally, PM014 treatment resulted in the downregulation of inflammatory cytokines, chemokines, and fibrosis-related genes and a reduction in the transforming growth factor-β1-positive cell population in lung tissue. Thus, PM014 is a potent therapeutic agent for radiation-induced lung fibrosis and inflammation.
Collapse
|
18
|
MicroRNA-Related Polymorphisms in PI3K/Akt/mTOR Pathway Genes Are Predictive of Limited-Disease Small Cell Lung Cancer Treatment Outcomes. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6501385. [PMID: 28280736 PMCID: PMC5322445 DOI: 10.1155/2017/6501385] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 11/30/2016] [Accepted: 12/15/2016] [Indexed: 01/05/2023]
Abstract
The phosphoinositide-3 kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway plays an important role in cancer progression and treatment, including that of small cell lung cancer (SCLC), a disease with traditionally poor prognosis. Given the regulatory role of microRNA (miRNA) in gene expression, we examined the association of single nucleotide polymorphisms (SNPs) at miRNA-binding sites of genes in the mTOR pathway with the prognosis of patients with limited-disease SCLC. A retrospective study was conducted of 146 patients with limited-disease SCLC treated with chemoradiotherapy. Nine SNPs of six mTOR pathway genes were genotyped using blood samples. Cox proportional hazard regression modeling and recursive partitioning analysis were performed to identify SNPs significantly associated with overall survival. Three SNPs, MTOR: rs2536 (T>C), PIK3R1: rs3756668 (A>G), and PIK3R1: rs12755 (A>C), were associated with longer overall survival. Recursive partitioning analysis based on unfavorable genotype combinations of the rs2536 and rs3756668 SNPs classified patients into three risk subgroups and was internally validated with 1000 bootstrap samples. These findings suggest that miRNA-related polymorphisms in the PI3K/Akt/mTOR pathway may be valuable biomarkers to complement clinicopathological variables in predicting prognosis of limited-disease SCLC and to facilitate selection of patients likely to benefit from chemoradiotherapy.
Collapse
|
19
|
Shin D, Lee G, Sohn SH, Park S, Jung KH, Lee JM, Yang J, Cho J, Bae H. Regulatory T Cells Contribute to the Inhibition of Radiation-Induced Acute Lung Inflammation via Bee Venom Phospholipase A₂ in Mice. Toxins (Basel) 2016; 8:131. [PMID: 27144583 PMCID: PMC4885046 DOI: 10.3390/toxins8050131] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/23/2016] [Accepted: 04/26/2016] [Indexed: 01/30/2023] Open
Abstract
Bee venom has long been used to treat various inflammatory diseases, such as rheumatoid arthritis and multiple sclerosis. Previously, we reported that bee venom phospholipase A₂ (bvPLA₂) has an anti-inflammatory effect through the induction of regulatory T cells. Radiotherapy is a common anti-cancer method, but often causes adverse effects, such as inflammation. This study was conducted to evaluate the protective effects of bvPLA₂ in radiation-induced acute lung inflammation. Mice were focally irradiated with 75 Gy of X-rays in the lung and administered bvPLA₂ six times after radiation. To evaluate the level of inflammation, the number of immune cells, mRNA level of inflammatory cytokine, and histological changes in the lung were measured. BvPLA₂ treatment reduced the accumulation of immune cells, such as macrophages, neutrophils, lymphocytes, and eosinophils. In addition, bvPLA₂ treatment decreased inflammasome-, chemokine-, cytokine- and fibrosis-related genes' mRNA expression. The histological results also demonstrated the attenuating effect of bvPLA₂ on radiation-induced lung inflammation. Furthermore, regulatory T cell depletion abolished the therapeutic effects of bvPLA₂ in radiation-induced pneumonitis, implicating the anti-inflammatory effects of bvPLA₂ are dependent upon regulatory T cells. These results support the therapeutic potential of bvPLA₂ in radiation pneumonitis and fibrosis treatments.
Collapse
Affiliation(s)
- Dasom Shin
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 130-701, South Korea.
| | - Gihyun Lee
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 130-701, South Korea.
| | - Sung-Hwa Sohn
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul 120-752, South Korea.
| | - Soojin Park
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 130-701, South Korea.
| | - Kyung-Hwa Jung
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 130-701, South Korea.
| | - Ji Min Lee
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul 120-752, South Korea.
| | - Jieun Yang
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 130-701, South Korea.
| | - Jaeho Cho
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul 120-752, South Korea.
| | - Hyunsu Bae
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 130-701, South Korea.
| |
Collapse
|
20
|
Pardo OE, Castellano L, Munro CE, Hu Y, Mauri F, Krell J, Lara R, Pinho FG, Choudhury T, Frampton AE, Pellegrino L, Pshezhetskiy D, Wang Y, Waxman J, Seckl MJ, Stebbing J. miR-515-5p controls cancer cell migration through MARK4 regulation. EMBO Rep 2016; 17:570-84. [PMID: 26882547 PMCID: PMC4818771 DOI: 10.15252/embr.201540970] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 12/22/2015] [Accepted: 01/13/2016] [Indexed: 01/28/2023] Open
Abstract
Here, we show that miR-515-5p inhibits cancer cell migration and metastasis. RNA-seq analyses of both oestrogen receptor receptor-positive and receptor-negative breast cancer cells overexpressing miR-515-5p reveal down-regulation of NRAS, FZD4, CDC42BPA, PIK3C2B and MARK4 mRNAs. We demonstrate that miR-515-5p inhibits MARK4 directly 3' UTR interaction and that MARK4 knock-down mimics the effect of miR-515-5p on breast and lung cancer cell migration. MARK4 overexpression rescues the inhibitory effects of miR-515-5p, suggesting miR-515-5p mediates this process through MARK4 down-regulation. Furthermore, miR-515-5p expression is reduced in metastases compared to primary tumours derived from both in vivo xenografts and samples from patients with breast cancer. Conversely, miR-515-5p overexpression prevents tumour cell dissemination in a mouse metastatic model. Moreover, high miR-515-5p and low MARK4 expression correlate with increased breast and lung cancer patients' survival, respectively. Taken together, these data demonstrate the importance of miR-515-5p/MARK4 regulation in cell migration and metastasis across two common cancers.
Collapse
Affiliation(s)
- Olivier E Pardo
- Department of Surgery & Cancer, Division of Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College Hammersmith Hospital Campus, London, UK
| | - Leandro Castellano
- Department of Surgery & Cancer, Division of Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College Hammersmith Hospital Campus, London, UK
| | - Catriona E Munro
- Department of Surgery & Cancer, Division of Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College Hammersmith Hospital Campus, London, UK
| | - Yili Hu
- Department of Oncology, Imperial College Healthcare NHS Trust Charing Cross Hospital, London, UK
| | - Francesco Mauri
- Department of Surgery & Cancer, Division of Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College Hammersmith Hospital Campus, London, UK
| | - Jonathan Krell
- Department of Surgery & Cancer, Division of Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College Hammersmith Hospital Campus, London, UK
| | - Romain Lara
- Department of Surgery & Cancer, Division of Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College Hammersmith Hospital Campus, London, UK
| | - Filipa G Pinho
- Department of Surgery & Cancer, Division of Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College Hammersmith Hospital Campus, London, UK
| | - Thameenah Choudhury
- Department of Surgery & Cancer, Division of Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College Hammersmith Hospital Campus, London, UK
| | - Adam E Frampton
- Department of Surgery & Cancer, Division of Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College Hammersmith Hospital Campus, London, UK
| | - Loredana Pellegrino
- Department of Surgery & Cancer, Division of Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College Hammersmith Hospital Campus, London, UK
| | - Dmitry Pshezhetskiy
- Department of Surgery & Cancer, Division of Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College Hammersmith Hospital Campus, London, UK
| | - Yulan Wang
- Department of Oncology, Imperial College Healthcare NHS Trust Charing Cross Hospital, London, UK
| | - Jonathan Waxman
- Department of Surgery & Cancer, Division of Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College Hammersmith Hospital Campus, London, UK
| | - Michael J Seckl
- Department of Surgery & Cancer, Division of Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College Hammersmith Hospital Campus, London, UK Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics Chinese Academy of Sciences, Wuhan, China
| | - Justin Stebbing
- Department of Surgery & Cancer, Division of Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College Hammersmith Hospital Campus, London, UK Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
21
|
Taylor MA, Wappett M, Delpuech O, Brown H, Chresta CM. Enhanced MAPK signaling drives ETS1-mediated induction of miR-29b leading to downregulation of TET1 and changes in epigenetic modifications in a subset of lung SCC. Oncogene 2016; 35:4345-57. [PMID: 26776158 PMCID: PMC4994018 DOI: 10.1038/onc.2015.499] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 11/16/2015] [Accepted: 11/20/2015] [Indexed: 01/21/2023]
Abstract
Non-small-cell lung cancer is the leading cause of cancer death worldwide and is comprised of several histological subtypes, the two most common being adenocarcinoma (AC) and squamous cell carcinoma (SCC). Targeted therapies have successfully improved response rates in patients with AC tumors. However, the majority of SCC tumors lack specific targetable mutations, making development of new treatment paradigms for this disease challenging. In the present study, we used iterative non-negative matrix factorization, an unbiased clustering method, on mRNA expression data from the cancer genome atlas (TCGA) and a panel of 24 SCC cell lines to classify three disease segments within SCC. Analysis of gene set enrichment and drug sensitivity identified an immune-evasion subtype that showed increased sensitivity to nuclear factor-κB and mitogen-activated protein kinase (MAPK) inhibition, a replication-stress associated subtype that showed increased sensitivity to ataxia telangiectasia inhibition, and a neuroendocrine-associated subtype that showed increased sensitivity to phosphoinositide 3-kinase and fibroblast growth factor receptor inhibition. Additionally, each of these subtypes exhibited a unique microRNA expression profile. Focusing on the immune-evasion subtype, bioinformatic analysis of microRNA promoters revealed enrichment for binding sites for the MAPK-driven ETS1 transcription factor. Indeed, we found that knockdown of ETS1 led to upregulation of eight microRNAs and downregulation of miR-29b in the immune-evasion subtype. Mechanistically, we found that miR-29b targets the DNA-demethylating enzyme, TET1, for downregulation resulting in decreased 5-hmC epigenetic modifications. Moreover, inhibition of MAPK signaling by gefitinib led to decreased ETS1 and miR-29b expression with a corresponding increase in TET1 expression and increase in 5-hmC. Collectively, our work identifies three subtypes of lung SCC that differ in drug sensitivity and shows a novel mechanism of miR-29b regulation by MAPK-driven ETS1 expression which leads to downstream changes in TET1-mediated epigenetic modifications.
Collapse
Affiliation(s)
- M A Taylor
- AstraZeneca, R&D Oncology iMed, Alderley Park, Macclesfield, UK
| | - M Wappett
- AstraZeneca, R&D Oncology iMed, Alderley Park, Macclesfield, UK
| | - O Delpuech
- AstraZeneca, R&D Oncology iMed, Alderley Park, Macclesfield, UK
| | - H Brown
- Personalised Healthcare and Biomarkers, AstraZeneca, Alderley Park, Macclesfield, UK
| | - C M Chresta
- AstraZeneca, R&D Oncology iMed, Alderley Park, Macclesfield, UK
| |
Collapse
|
22
|
Mura M, Hopkins TG, Michael T, Abd-Latip N, Weir J, Aboagye E, Mauri F, Jameson C, Sturge J, Gabra H, Bushell M, Willis AE, Curry E, Blagden SP. LARP1 post-transcriptionally regulates mTOR and contributes to cancer progression. Oncogene 2015; 34:5025-36. [PMID: 25531318 PMCID: PMC4430325 DOI: 10.1038/onc.2014.428] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 09/20/2014] [Accepted: 10/21/2014] [Indexed: 12/24/2022]
Abstract
RNA-binding proteins (RBPs) bind to and post-transcriptionally regulate the stability of mRNAs. La-related protein 1 (LARP1) is a conserved RBP that interacts with poly-A-binding protein and is known to regulate 5'-terminal oligopyrimidine tract (TOP) mRNA translation. Here, we show that LARP1 is complexed to 3000 mRNAs enriched for cancer pathways. A prominent member of the LARP1 interactome is mTOR whose mRNA transcript is stabilized by LARP1. At a functional level, we show that LARP1 promotes cell migration, invasion, anchorage-independent growth and in vivo tumorigenesis. Furthermore, we show that LARP1 expression is elevated in epithelial cancers such as cervical and non-small cell lung cancers, where its expression correlates with disease progression and adverse prognosis, respectively. We therefore conclude that, through the post-transcriptional regulation of genes such as mTOR within cancer pathways, LARP1 contributes to cancer progression.
Collapse
Affiliation(s)
- M Mura
- Division of Cancer, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Campus, London, UK
| | - T G Hopkins
- Division of Cancer, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Campus, London, UK
| | - T Michael
- Division of Cancer, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Campus, London, UK
| | - N Abd-Latip
- Division of Cancer, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Campus, London, UK
| | - J Weir
- Department of Cellular Pathology, Imperial College Healthcare NHS Trust, Charing Cross Hospital, London, UK
| | - E Aboagye
- Division of Cancer, Department of Surgery and Cancer, Cancer Research UK Laboratories, Imperial College London, Hammersmith Campus, London, UK
| | - F Mauri
- Department of Histopathology, Centre for Pathology, Imperial College London, Hammersmith Campus, London, UK
| | - C Jameson
- Department of Histopathology, University College Hospital, London, UK
| | - J Sturge
- Division of Cancer, Department of Surgery and Cancer, Cancer Research UK Laboratories, Imperial College London, Hammersmith Campus, London, UK
- School of Biological, Biomedical & Environmental Sciences, The Allam Building, University of Hull, Hull, UK
| | - H Gabra
- Division of Cancer, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Campus, London, UK
| | - M Bushell
- MRC Toxicology Unit, Hodgkin Building, University of Leicester, Leicester, UK
| | - A E Willis
- MRC Toxicology Unit, Hodgkin Building, University of Leicester, Leicester, UK
| | - E Curry
- Division of Cancer, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Campus, London, UK
| | - S P Blagden
- Division of Cancer, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Campus, London, UK
| |
Collapse
|
23
|
O'Leary K, Shia A, Cavicchioli F, Haley V, Comino A, Merlano M, Mauri F, Walter K, Lackner M, Wischnewsky MB, Crook T, Lo Nigro C, Schmid P. Identification of Endoglin as an epigenetically regulated tumour-suppressor gene in lung cancer. Br J Cancer 2015; 113:970-8. [PMID: 26325105 PMCID: PMC4578092 DOI: 10.1038/bjc.2015.302] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 07/17/2015] [Accepted: 07/29/2015] [Indexed: 12/25/2022] Open
Abstract
Background: The transforming growth factor-beta (TGF- β) pathway has been implicated in proliferation, migration and invasion of various cancers. Endoglin is a TGF-β accessory receptor that modulates signalling. We identified Endoglin as an epigenetically silenced tumour-suppressor gene in lung cancer by means of a genome-wide screening approach, then sought to characterise its effect on lung cancer progression. Methods: Methylation microarray and RNA sequencing were carried out on lung cancer cell lines. Epigenetic silencing of Endoglin was confirmed by methylation and expression analyses. An expression vector and a 20-gene expression panel were used to evaluate Endoglin function. Pyrosequencing was carried out on two independent cohorts comprising 112 and 202 NSCLC cases, respectively, and the impact of Endoglin methylation on overall survival (OS) was evaluated. Results: Methylation in the promoter region resulted in silencing of Endoglin, which could be reactivated by demethylation. Increased invasion coupled with altered EMT marker expression was observed in cell lines with an epithelial-like, but not those with a mesenchymal-like, profile when Endoglin was absent. Methylation was associated with decreased OS in stage I but not in stages II–III disease. Conclusions: We show that Endoglin is a common target of epigenetic silencing in lung cancer. We reveal a link between Endoglin silencing and EMT progression that might be associated with decreased survival in stage I disease.
Collapse
Affiliation(s)
- K O'Leary
- Brighton and Sussex Medical School, University of Sussex, Brighton BN1 9RY, UK
| | - A Shia
- Brighton and Sussex Medical School, University of Sussex, Brighton BN1 9RY, UK.,Barts Cancer Institute, Queen Mary University of London, Old Anatomy Building, Charterhouse Square, London EC1M 6BQ, UK
| | - F Cavicchioli
- Brighton and Sussex Medical School, University of Sussex, Brighton BN1 9RY, UK
| | - V Haley
- Brighton and Sussex Medical School, University of Sussex, Brighton BN1 9RY, UK
| | - A Comino
- Pathology Department, S. Croce General Hospital, via Coppino 26, 12100, Cuneo, Italy
| | - M Merlano
- Medical Oncology, Oncology Department, S. Croce General Hospital, via Carle 25, 12100, Cuneo, Italy
| | - F Mauri
- Department of Histopathology, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - K Walter
- Oncology Biomarker Development, Genentech, Inc., 550 Grandview Boulevard, South San Francisco, CA 94080, USA
| | - M Lackner
- Oncology Biomarker Development, Genentech, Inc., 550 Grandview Boulevard, South San Francisco, CA 94080, USA
| | - M B Wischnewsky
- eScience Lab, Department of Biomathematics, University of Bremen, Bremen 28359, Germany
| | - T Crook
- Division of Cancer Research, Medical Research Institute, Jacqui Wood Cancer Centre, University of Dundee, Ninewells Hospital And Medical School, Dundee DD1 9SY, UK
| | - C Lo Nigro
- Laboratory of Cancer Genetics and Translational Oncology, Oncology Department, S. Croce Genreal Hospital, via Carle 25, Cuneo 12100, Italy
| | - P Schmid
- Brighton and Sussex Medical School, University of Sussex, Brighton BN1 9RY, UK.,Barts Cancer Institute, Queen Mary University of London, Old Anatomy Building, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
24
|
Shin E, Choi CM, Kim HR, Jang SJ, Park YS. Immunohistochemical characterization of the mTOR pathway in stage-I non-small-cell lung carcinoma. Lung Cancer 2015; 89:13-8. [PMID: 25936472 DOI: 10.1016/j.lungcan.2015.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 03/25/2015] [Accepted: 04/02/2015] [Indexed: 01/17/2023]
Abstract
BACKGROUND Dysregulation of mammalian target of rapamycin (mTOR) pathway has been linked with malignant tumorigenesis. This study explored the expression profiles of proteins involved in the mTOR pathway and their relationships with clinicopathologic characteristics in stage-I non-small-cell lung carcinoma (NSCLC). METHODS The protein expression profiles of PTEN, p-Akt, p-mTOR, p-S6, and eIF4E were examined using immunohistochemical staining and tissue microarray method in 408 patients with stage-I NSCLC (250 adenocarcinomas [ADC] and 158 squamous cell carcinomas). RESULTS Retained PTEN expression (P<0.001), p-mTOR expression (P<0.001), and p-S6 expression (P=0.007) were associated with ADC histology. Expression of PTEN (P=0.001), p-Akt (P=0.005), p-mTOR (P=0.007), p-S6 (P<0.001) were correlated with lower pathologic T stage. PTEN loss was correlated with male gender and smoking history and p-mTOR expression was inversely correlated with these factors (P<0.001). Subgroup analysis of ADCs indicated that male gender, high pT stage, lymphovascular invasion, and PTEN loss were poor prognostic factors. Multivariate analysis revealed that the PTEN(-)/p-Akt(+)/p-mTOR(+) combination more effectively determined the prognosis of ADC (hazard ratio=2.2, P=0.004) than PTEN alone. CONCLUSIONS Activation of the mTOR pathway in early-stage ADCs suggests a significant role for the mTOR axis in early carcinogenesis. The combination of PTEN(-)/p-Akt(+)/p-mTOR(+) expression was correlated with poor overall survival in patients with stage-I lung ADC.
Collapse
Affiliation(s)
- Eun Shin
- Department of Pathology, Seoul National University Bundang Hospital, 173-82 Gumiro, Bundang-gu, Seongnam 463-707, Gyeonggi-do, Republic of Korea
| | - Chang Min Choi
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, Songpa-Gu, 138-736 Seoul, Republic of Korea
| | - Hyeong Ryul Kim
- Department of Thoracic Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, Songpa-Gu, 138-736 Seoul, Republic of Korea
| | - Se Jin Jang
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, Songpa-Gu, 138-736 Seoul, Republic of Korea
| | - Young Soo Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, Songpa-Gu, 138-736 Seoul, Republic of Korea.
| |
Collapse
|
25
|
Sohn SH, Lee JM, Park S, Yoo H, Kang JW, Shin D, Jung KH, Lee YS, Cho J, Bae H. The inflammasome accelerates radiation-induced lung inflammation and fibrosis in mice. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 39:917-926. [PMID: 25805627 DOI: 10.1016/j.etap.2015.02.019] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 02/24/2015] [Accepted: 02/27/2015] [Indexed: 06/04/2023]
Abstract
Although lung inflammation and fibrosis are well-documented dose-limiting side effects of lung irradiation, the mechanisms underlying these pathologies are unknown. An improved mechanistic understanding of radiation-induced pneumonitis is a prerequisite for the development of more effective radiotherapy; this was the rationale for the current study. Mouse lungs were focally irradiated with 75 Gy. The numbers of neutrophils, lymphocytes, macrophages, and total cells in the bronchoalveolar lavage fluid were counted, and pro-inflammatory cytokine levels were measured. Histological analysis and immunohistochemical staining for Tgf-β1 and Cd68 (a macrophage-specific protein) was also performed. After irradiation, mice developed pneumonitis, and exhibited higher numbers of neutrophils, lymphocytes, eosinophils, macrophages, and total cells compared to controls. In addition, inflammasome (Nlrp3, and caspase 1, Il1a, and Il1β), adhesion molecule (Vcam1), and cytokine (Il6) genes were significantly upregulated in the IR group. Cd68 and Tgfb1 proteins were significantly increased after irradiation. Upregulation of Cd68 and Tgfb1 correlates with the onset of radiation-induced pneumonitis and fibrosis. In addition, radiation-induced pneumonitis and fibrosis are accompanied by upregulation of phenotypic markers of inflammasome activity. Our findings have implications for the onset and exacerbation of damage in normal lung tissue.
Collapse
Affiliation(s)
- Sung-Hwa Sohn
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji Min Lee
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, South Korea
| | - Soojin Park
- Department of Physiology, College of Oriental Medicine, Kyung Hee University, Seoul, South Korea
| | - Hyun Yoo
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, South Korea
| | - Jeong Wook Kang
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, South Korea
| | - Dasom Shin
- Department of Physiology, College of Oriental Medicine, Kyung Hee University, Seoul, South Korea
| | - Kyung-Hwa Jung
- Department of Physiology, College of Oriental Medicine, Kyung Hee University, Seoul, South Korea
| | - Yun-Sil Lee
- College of Pharmacy & Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, South Korea
| | - Jaeho Cho
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, South Korea.
| | - Hyunsu Bae
- Department of Physiology, College of Oriental Medicine, Kyung Hee University, Seoul, South Korea.
| |
Collapse
|
26
|
Li L, Liu D, Qiu ZX, Zhao S, Zhang L, Li WM. The prognostic role of mTOR and p-mTOR for survival in non-small cell lung cancer: a systematic review and meta-analysis. PLoS One 2015; 10:e0116771. [PMID: 25680114 PMCID: PMC4332670 DOI: 10.1371/journal.pone.0116771] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 12/13/2014] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES The mammalian target of rapamycin (mTOR) and phosphorylated mTOR (p-mTOR) are potential prognostic markers and therapeutic targets for non-small cell lung cancer (NSCLC). However, the association between mTOR/p-mTOR expression and NSCLC patients' prognosis remains controversial. Thus, a meta-analysis of existing studies evaluating the prognostic role of mTOR/p-mTOR expression for NSCLC was conducted. MATERIALS AND METHODS A systemically literature search was performed via Pubmed, Embase, Medline as well as CNKI (China National Knowledge Infrastructure). Studies were included that reported the hazard ratio (HR) and 95%CI for the association between mTOR/p-mTOR expression and NSCLC patients' survival. Random-effects model was used to pool HRs. RESULTS Ten eligible studies were included in this meta-analysis, with 4 about m-TOR and 7 about p-mTOR. For mTOR, the pooled HR of overall survival (OS) was 1.00 (95%CI 0.5 to 1.99) by univariate analysis and 1.22 (95%CI 0.53 to 2.82) by multivariate analysis. For p-mTOR, the pooled HR was 1.39 (95%CI 0.97 to 1.98) by univariate analysis and 1.42 (95%CI 0.56 to 3.60) by multivariate analysis. CONCLUSION The results indicated that no statistically significant association was found between mTOR/p-mTOR expression and NSCLC patients' prognosis.
Collapse
Affiliation(s)
- Lei Li
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Dan Liu
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Zhi-Xin Qiu
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Shuang Zhao
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Li Zhang
- Lab of Pathology, Department of Pathology, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Wei-Min Li
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| |
Collapse
|
27
|
Roy R, Durie D, Li H, Liu BQ, Skehel JM, Mauri F, Cuorvo LV, Barbareschi M, Guo L, Holcik M, Seckl MJ, Pardo OE. hnRNPA1 couples nuclear export and translation of specific mRNAs downstream of FGF-2/S6K2 signalling. Nucleic Acids Res 2014; 42:12483-97. [PMID: 25324306 PMCID: PMC4227786 DOI: 10.1093/nar/gku953] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/26/2014] [Accepted: 09/29/2014] [Indexed: 01/14/2023] Open
Abstract
The increased cap-independent translation of anti-apoptotic proteins is involved in the development of drug resistance in lung cancer but signalling events regulating this are poorly understood. Fibroblast growth factor 2 (FGF-2) signalling-induced S6 kinase 2 (S6K2) activation is necessary, but the downstream mediator(s) coupling this kinase to the translational response is unknown. Here, we show that S6K2 binds and phosphorylates hnRNPA1 on novel Ser4/6 sites, increasing its association with BCL-XL and XIAP mRNAs to promote their nuclear export. In the cytoplasm, phosphoS4/6-hnRNPA1 dissociates from these mRNAs de-repressing their IRES-mediated translation. This correlates with the phosphorylation-dependent association of hnRNPA1 with 14-3-3 leading to hnRNPA1 sumoylation on K183 and its re-import into the nucleus. A non-phosphorylatible, S4/6A mutant prevented these processes, hindering the pro-survival activity of FGF-2/S6K2 signalling. Interestingly, immunohistochemical staining of lung and breast cancer tissue samples demonstrated that increased S6K2 expression correlates with decreased cytoplasmic hnRNPA1 and increased BCL-XL expression. In short, phosphorylation on novel N-term sites of hnRNPA1 promotes translation of anti-apoptotic proteins and is indispensable for the pro-survival effects of FGF-2.
Collapse
Affiliation(s)
- Rajat Roy
- Division of Cancer, Department of Surgery and Cancer, 1st Floor, ICTEM Building, Hammersmith Hospitals Campus of Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Danielle Durie
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Hui Li
- Department of Biochemistry, Wuhan University, Wuhan, China
| | - Bing-Qian Liu
- Department of Biochemistry, Wuhan University, Wuhan, China
| | - John Mark Skehel
- Protein Analysis and Proteomics Laboratory, London Research Institute, South Mimms, EN6 3LD, UK
| | - Francesco Mauri
- Department of Histopathology, Hammersmith Hospital Campus, Imperial College, London W120NN, UK
| | | | | | - Lin Guo
- Department of Biochemistry, Wuhan University, Wuhan, China
| | - Martin Holcik
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Michael J Seckl
- Division of Cancer, Department of Surgery and Cancer, 1st Floor, ICTEM Building, Hammersmith Hospitals Campus of Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Olivier E Pardo
- Division of Cancer, Department of Surgery and Cancer, 1st Floor, ICTEM Building, Hammersmith Hospitals Campus of Imperial College London, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
28
|
Yasugi M, Takigawa N, Ochi N, Ohashi K, Harada D, Ninomiya T, Murakami T, Honda Y, Ichihara E, Tanimoto M, Kiura K. Everolimus prolonged survival in transgenic mice with EGFR-driven lung tumors. Exp Cell Res 2014; 326:201-9. [PMID: 24768699 DOI: 10.1016/j.yexcr.2014.04.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 04/02/2014] [Accepted: 04/14/2014] [Indexed: 12/14/2022]
Abstract
Everolimus is an orally administered mTOR inhibitor. The effect, and mechanism of action, of everolimus on lung cancers with an epidermal growth factor receptor (EGFR) mutation remain unclear. Four gefitinib-sensitive and -resistant cell lines were used in the present work. Growth inhibition was determined using the MTT assay. Transgenic mice carrying the EGFR L858R mutation were treated with everolimus (10 mg/kg/day), or vehicle alone, from 5 to 20 weeks of age, and were then sacrificed. To evaluate the efficacy of everolimus in prolonging survival, everolimus (10 mg/kg/day) or vehicle was administered from 5 weeks of age. The four cell lines were similarly sensitive to everolimus. Expression of phosphorylated (p) mTOR and pS6 were suppressed upon treatment with everolimus in vitro, whereas the pAKT level increased. The numbers of lung tumors with a long axis exceeding 1mm in the everolimus-treated and control groups were 1.9 ± 0.9 and 9.4 ± 3.2 (t-test, p<0.001), respectively. pS6 was suppressed during eve r olimus treatment. Although apoptosis and autophagy were not induced in everolimus-treated EGFR transgenic mice, angiogenesis was suppressed. The median survival time in the everolimus-treated group (58.0 weeks) was significantly longer than that in the control group (31.2 weeks) (logrank test, p<0.001). These findings suggest that everolimus had an indirect effect on tumor formation by inhibiting angiogenesis and might be effective to treat lung tumors induced by an activating EGFR gene mutation.
Collapse
Affiliation(s)
- Masayuki Yasugi
- Department of Hematology, Oncology, and Respiratory Medicine, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences and Okayama University Hospital, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Nagio Takigawa
- Department of General Internal Medicine 4, Kawasaki Hospital, Kawasaki Medical School, Okayama 700-8505, Japan.
| | - Nobuaki Ochi
- Department of Hematology, Oncology, and Respiratory Medicine, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences and Okayama University Hospital, 2-5-1 Shikata-cho, Okayama 700-8558, Japan; Department of General Internal Medicine 4, Kawasaki Hospital, Kawasaki Medical School, Okayama 700-8505, Japan
| | - Kadoaki Ohashi
- Department of Hematology, Oncology, and Respiratory Medicine, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences and Okayama University Hospital, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Daijiro Harada
- Department of Hematology, Oncology, and Respiratory Medicine, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences and Okayama University Hospital, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Takashi Ninomiya
- Department of Hematology, Oncology, and Respiratory Medicine, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences and Okayama University Hospital, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Toshi Murakami
- Department of Hematology, Oncology, and Respiratory Medicine, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences and Okayama University Hospital, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Yoshihiro Honda
- Department of Hematology, Oncology, and Respiratory Medicine, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences and Okayama University Hospital, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Eiki Ichihara
- Department of Hematology, Oncology, and Respiratory Medicine, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences and Okayama University Hospital, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Mitsune Tanimoto
- Department of Hematology, Oncology, and Respiratory Medicine, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences and Okayama University Hospital, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Katsuyuki Kiura
- Department of Hematology, Oncology, and Respiratory Medicine, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences and Okayama University Hospital, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| |
Collapse
|
29
|
Pinato DJ, Shiner RJ, Seckl MJ, Stebbing J, Sharma R, Mauri FA. Prognostic performance of inflammation-based prognostic indices in primary operable non-small cell lung cancer. Br J Cancer 2014; 110:1930-5. [PMID: 24667648 PMCID: PMC3992503 DOI: 10.1038/bjc.2014.145] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 02/11/2014] [Accepted: 02/24/2014] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND At least 30% of patients with primary resectable non-small cell lung cancer (NSCLC) will experience a relapse in their disease within 5 years following definitive treatment. Clinicopathological predictors have proved to be suboptimal in identifying high-risk patients. We aimed to establish whether inflammation-based scores offer an improved prognostic ability in terms of estimating overall (OS) and recurrence-free survival (RFS) in a cohort of operable, early-stage NSCLC patients. METHODS Clinicopathological, demographic and treatment data were collected prospectively for 220 patients operated for primary NSCLC at the Hammersmith Hospital from 2004 to 2011. Pretreatment modified Glasgow Prognostic Score (mGPS), neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) were tested together with established prognostic factors in uni- and multivariate Cox regression analyses of OS and RFS. RESULTS Half of the patients were male, with a median age of 65. A total of 57% were classified as stage I with adenocarcinoma being the most prevalent subtype (60%). Univariate analyses of survival revealed stage (P<0.001), grade (P=0.02), lymphovascular (LVI, P=0.001), visceral pleural invasion (VPI, P=0.003), mGPS (P=0.02) and NLR (P=0.04) as predictors of OS, with stage (P<0.001), VPI (P=0.02) and NLR (P=0.002) being confirmed as independent prognostic factors on multivariate analyses. Patients with more advanced stage (P<0.001) and LVI (P=0.008) had significantly shorter RFS. CONCLUSIONS An elevated NLR identifies operable NSCLC patients with a poor prognostic outlook and an OS difference of almost 2 years compared to those with a normal score at diagnosis. Our study validates the clinical utility of the NLR in early-stage NSCLC.
Collapse
Affiliation(s)
- D J Pinato
- Division of Experimental Medicine, Hammersmith Campus of Imperial College London, Du Cane Road, W120HS London, UK
| | - R J Shiner
- Department of Respiratory Medicine, National Heart and Lung Institute, Hammersmith Hospital, Du Cane Road, W120HS London, UK
| | - M J Seckl
- Division of Oncology, Hammersmith Campus of Imperial College London, Du Cane Road, W120HS London, UK
| | - J Stebbing
- Division of Oncology, Hammersmith Campus of Imperial College London, Du Cane Road, W120HS London, UK
| | - R Sharma
- Division of Experimental Medicine, Hammersmith Campus of Imperial College London, Du Cane Road, W120HS London, UK
| | - F A Mauri
- Department of Pathology, Hammersmith Campus of Imperial College London, Du Cane Road, W120HS London, UK
| |
Collapse
|
30
|
Tuberous-sclerosis complex-related cell signaling in the pathogenesis of lung cancer. Diagn Pathol 2014; 9:48. [PMID: 24593867 PMCID: PMC3975884 DOI: 10.1186/1746-1596-9-48] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 02/22/2014] [Indexed: 12/12/2022] Open
Abstract
Background Hamartin (TSC1) and tuberin (TSC2), encoded by the tuberous sclerosis complex (TSC) genes, form a tumor-suppressor heterodimer which is implicated in PI3K-Akt signaling and acts as a functional inhibitor of the mammalian target of rapamycin (mTOR). Dysregulation of mTOR has been assigned to carcinogenesis and thus may be involved in cancer development. We have addressed the role of hamartin, phospho-tuberin (p-TSC2) and phospho-mTOR (p-mTOR) in a series of non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) samples. Methods We collected 166 NSCLC and SCLC samples for immunohistochemical studies and performed western blot analyses in NSCLC and SCLC cell lines as well as comparative analyses with EGFR phosphorylation and downstream effectors. Results In cell lines we found an inverse correlation between hamartin and p-mTOR expression. In surgical specimens cytoplasmic hamartin expression was observed in more than 50% of adenocarcinoma (AC) and squamous cell carcinoma (SCC) compared to 14% of SCLC. P-mTOR and p-TSC2 staining was found in a minority of cases. There was a significant correlation between p-EGFR Tyr-1068, p-EGFR Tyr-992 and hamartin, and also between p-mTOR and p-EGFR Tyr-1173 in AC. In SCC an inverse correlation between hamartin and p-EGFR Tyr-992 was detected. Phosphorylation of TSC2 was associated with expression of MAP-Kinase. Hamartin, p-TSC2 and p-mTOR expression was not dependant of the EGFR mutation status. Hamartin expression is associated with poorer survival in SCC and SCLC. Conclusions Our findings confirm the inhibitory role of the tuberous sclerosis complex for mTOR activation in lung cancer cell lines. These results reveal hamartin expression in a substantial subset of NSCLC and SCLC specimens, which may be due to EGFR signaling but is not dependant on EGFR mutations. Our data provide evidence for a functional role of the tuberous sclerosis complex in lung cancer. Virtual slides The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/9274845161175223.
Collapse
|
31
|
TRIGKA ELENIANDRIANA, LEVIDOU GEORGIA, SAETTA ANGELICAA, CHATZIANDREOU ILENIA, TOMOS PERIKLIS, THALASSINOS NIKOLAOS, ANASTASIOU NIKOLAOS, SPARTALIS ELEFTHERIOS, KAVANTZAS NIKOLAOS, PATSOURIS EFSTRATIOS, KORKOLOPOULOU PENELOPE. A detailed immunohistochemical analysis of the PI3K/AKT/mTOR pathway in lung cancer: Correlation with PIK3CA, AKT1, K-RAS or PTEN mutational status and clinicopathological features. Oncol Rep 2013; 30:623-36. [DOI: 10.3892/or.2013.2512] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 02/08/2013] [Indexed: 11/06/2022] Open
|
32
|
Abstract
BACKGROUND An understanding of the activated protein signaling architecture in non-small-cell lung cancer (NSCLC) is of critical importance to the development of new therapeutic approaches and identification of predictive and prognostic biomarkers for patient stratification. METHODS We used reverse-phase protein microarrays to map the activated protein signaling networks of 47 NSCLC tumors, 28 of which were node negative, which were subjected to tumor cellular enrichment using laser capture microdissection. The phosphorylation/cleavage levels of 111 key signaling proteins and total levels of 17 proteins were measured for broadscale signaling analysis. RESULTS Pathway activation mapping of NSCLC revealed distinct subgroups composed of epidermal growth factor receptor (ERBB1), v-erb-b2 erythroblastic leukemia viral oncogene homolog 2 (ERBB2), v-erb-b2 erythroblastic leukemia viral oncogene homolog 3 (ERBB3), v-erb-a erythroblastic leukemia viral oncogene homolog 4 (ERBB4), v-akt murine thymoma viral oncogene homolog 1- mammalian target of rapamycin (AKT-mTOR), protein kinase, AMP-activated, alpha 2 catalytic subunit (AMPK), and autophagy-related signaling, along with transforming growth factor-beta-signaling protein 1 (SMAD), insulin-line growth factor receptor (IGFR), rearranged during transfection proto-oncogene (RET), and activated CDC42-associated kinase (ACK) activation. Investigation of epidermal growth factor receptor (EGFR)-driven signaling identified a unique cohort of tumors with low EGFR protein expression yet high relative levels of phosphorylated EGFR and high EGFR total protein with low relative levels of phosphorylation. Last, mapping analysis of patients with NSCLC with N0 disease revealed a pilot pathway activation signature composed of linked epidermal growth factor receptor family (HER)-AMPK-AKT-mTOR signaling network along with focal adhesion kinase- LIM domain kinase-1 (FAK-LIMK) and janus kinase (JAK)-signal transducers and activators of transcription (STAT) pathways that correlated with short-term survival and aggressive disease. CONCLUSIONS Functional protein pathway activation mapping of NSCLC reveals distinct activation subgroups that are underpinned by important therapeutic targets and that patients with early-stage node negative disease and poor prognosis may be identified by activation of defined, biochemically linked protein signaling events. Such findings, if confirmed in larger study sets, could help select and stratify patients for personalized targeted therapies.
Collapse
|
33
|
Chung JY, Kitano H, Takikita M, Cho H, Noh KH, Kim TW, Ylaya K, Hanaoka J, Fukuoka J, Hewitt SM. Synaptonemal complex protein 3 as a novel prognostic marker in early stage non-small cell lung cancer. Hum Pathol 2013; 44:472-9. [PMID: 23069255 PMCID: PMC3548971 DOI: 10.1016/j.humpath.2012.06.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 06/22/2012] [Accepted: 06/27/2012] [Indexed: 12/25/2022]
Abstract
Synaptonemal complex protein 3 is a marker for cell transformation that has prognostic significance in various cancers. However, the prognostic significance of synaptonemal complex protein 3 has not been studied in non-small cell lung cancer. To investigate the potential correlation between synaptonemal complex protein 3 and various clinicopathologic parameters, we assessed the expression of synaptonemal complex protein 3 in archival tumor tissues from 258 patients with non-small cell lung cancer by immunohistochemical staining. By immunofluorescence, synaptonemal complex protein 3 was detected in both the cytoplasmic and nuclear fractions of NCI-H1299 cell. In tumor samples, synaptonemal complex protein 3 is detected as cytoplasmic expression pattern and observed in 50 clinical samples (19.4%) by immunohistochemical staining. Synaptonemal complex protein 3 expression was correlated with T status (P = .008), lymph node metastasis (P = .010), tumor types (P = .019), and pleural invasion (P = .005). In multivariate analysis of patients with early stage disease, increased synaptonemal complex protein 3 expression predicted worse overall survival in early stage (stage I and II) with pT1 status (P = .041). These results suggest that positive synaptonemal complex protein 3 expression is a portent of poor outcome and may be a potential biomarker in the early stages of the non-small cell lung cancer for survival and may provide clues in the identification of patients for adjuvant therapy.
Collapse
Affiliation(s)
- Joon-Yong Chung
- Tissue Array Research Program & Applied Molecular Pathology Laboratory, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Haruhisa Kitano
- Tissue Array Research Program & Applied Molecular Pathology Laboratory, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Thoracic Surgery, Shiga University of Medical Science, Otsu, Japan
| | - Mikiko Takikita
- Tissue Array Research Program & Applied Molecular Pathology Laboratory, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Division of Diagnostic Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Hanbyoul Cho
- Tissue Array Research Program & Applied Molecular Pathology Laboratory, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kyung Hee Noh
- Department of Biomedical Sciences, Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Tae Woo Kim
- Department of Biomedical Sciences, Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Kris Ylaya
- Tissue Array Research Program & Applied Molecular Pathology Laboratory, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jun Hanaoka
- Department of Thoracic Surgery, Shiga University of Medical Science, Otsu, Japan
| | - Junya Fukuoka
- Laboratory of Pathology, Toyama University Hospital, Toyama, Japan
| | - Stephen M. Hewitt
- Tissue Array Research Program & Applied Molecular Pathology Laboratory, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
34
|
Zhang Z, Wang M. [Advanced research of mTOR and lung carcinoid tumors]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2013; 16:43-7. [PMID: 23327873 PMCID: PMC6000455 DOI: 10.3779/j.issn.1009-3419.2013.01.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Revised: 12/12/2012] [Indexed: 11/25/2022]
Abstract
Mammalian target of rapamycin (mTOR), a main protein kinase in the phosphoinositide 3-kinase (PI3K)/AKT/mTOR signaling pathway, is an important intracellular mediator involved in multiple celluar functions including proliferation, differentiation, apoptosis, tumorigenesis, and angiogenesis. Recently, the high expression of mTOR and mTOR-related kinase have been found in neuroendocrin tumors. Therefore, mTOR pathway represents an attractive target for new anticancer therapies except surgery.
Collapse
Affiliation(s)
- Zixuan Zhang
- Department of Respiratory Diseases, Peking Union Medical college Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | | |
Collapse
|
35
|
Immunohistochemical markers of the hypoxic response can identify malignancy in phaeochromocytomas and paragangliomas and optimize the detection of tumours with VHL germline mutations. Br J Cancer 2012; 108:429-37. [PMID: 23257898 PMCID: PMC3566818 DOI: 10.1038/bjc.2012.538] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: There are no reliable markers of malignancy in phaeochromocytomas (PCC) and paragangliomas (PGL). We investigated the relevance of the mammalian target of rapamycin (mTOR)/AKT and hypoxic pathways as novel immunohistochemical markers of malignancy. Methods: Tissue microarray blocks were constructed with a total of 100 tumours (10 metastatic) and 20 normal adrenomedullary samples. Sections were immunostained for hypoxia-inducible factor 1α (Hif-1α), vascular endothelial growth factor A (VEGF-A), mTOR, carbonic anhydrase IX (CaIX) and AKT. The predictive performance of these markers was studied using univariate, multivariate and receiver operating characteristic analyses. Results: In all, 100 consecutive patients, 64% PCC, 29% familial with a median tumour size of 4.7 cm (range 1–14) were included. Univariate analyses showed Hif-1α overexpression, tumour necrosis, size >5 cm, capsular and vascular invasion to be predictors of metastasis. In multivariate analysis, Hif-1α, necrosis and vascular invasion remained as independent predictors of metastasis. Hif-1α was the most discriminatory biomarker for the presence of metastatic diffusion. Strong membranous CaIX expression was seen in von Hippel–Lindau (VHL) PCC as opposed to other subtypes. Conclusion: Lack of vascular invasion, tumour necrosis and low Hif-1α expression identify tumours with lower risk of malignancy. We propose membranous CaIX expression as a potential marker for VHL disease in patients presenting with PCC.
Collapse
|
36
|
Qiu ZX, Wang L, Han J, Liu D, Huang W, Altaf K, Qiu XS, Javed MA, Zheng J, Chen BJ, Li WM. Prognostic impact of Raf-1 and p-Raf-1 expressions for poor survival rate in non-small cell lung cancer. Cancer Sci 2012; 103:1774-9. [PMID: 22738312 DOI: 10.1111/j.1349-7006.2012.02375.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 06/13/2012] [Accepted: 06/23/2012] [Indexed: 12/21/2022] Open
Abstract
Overexpression of Raf-1 has commonly been observed in solid tumors including non-small cell lung cancer (NSCLC). The objective of this study was to investigate whether overexpression of Raf-1, phosphorylated-Raf-1 (p-Raf-1) or both correlates with poor survival rate in NSCLC patients and to explore associations between expression of these proteins and NSCLC cell fate both in vitro and in vivo. Expression of Raf-1 and p-Raf-1 were detected by immunohistochemistry in tumor specimens from 152 NSCLC patients and associations between their expression and the clinicopathological characteristics were assessed. Five-year median survival rate of patients were analyzed by Kaplan-Meier method, log-rank test and Cox regression. Cell fate was compared between normal tumor cells and those with Raf-1 silencing, in both the adenocarcinoma cell line A549 and xenografted mice that were infected with the A549 cell line. The incidence of overexpression of both Raf-1 and p-Raf-1 in NSCLC was much higher than normal control (P < 0.05), and the survival rate of patients with positive expression of Raf-1, p-Raf-1 or both was found to be significantly lower than the negative group (P < 0.05). Both univariate and multivariate analyses showed Raf-1 (P = 0.000, P = 0.010), p-Raf-1 (P = 0.004, P = 0.046), or both (P = 0.001, P = 0.016) was good prognostic markers for poor survival rate in NSCLC patients. Suppression of Raf-1 inhibited tumorigenesis by inducing apoptosis both in vitro and in vivo. These findings demonstrate that overexpression of Raf-1, p-Raf-1 or both could be considered as a new independent prognostic biomarker for poor survival rates for NSCLC patients.
Collapse
Affiliation(s)
- Zhi-Xin Qiu
- Department of Respiratory Medicine, Sichuan University, Chengdu, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
MAURI FRANCESCOA, PINATO DAVIDJ, TRIVEDI PRITESH, SHARMA ROHINI, SHINER ROBERTJ. Isogeneic comparison of primary and metastatic lung cancer identifies CX3CR1 as a molecular determinant of site-specific metastatic diffusion. Oncol Rep 2012; 28:647-53. [DOI: 10.3892/or.2012.1818] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 04/30/2012] [Indexed: 11/06/2022] Open
|
38
|
Oh MH, Lee HJ, Yoo SB, Xu X, Choi JS, Kim YH, Lee SY, Lee CT, Jheon S, Chung JH. Clinicopathological correlations of mTOR and pAkt expression in non-small cell lung cancer. Virchows Arch 2012; 460:601-9. [PMID: 22562131 DOI: 10.1007/s00428-012-1239-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2012] [Accepted: 04/16/2012] [Indexed: 12/26/2022]
Abstract
The Akt/mammalian target of rapamycin (mTOR) pathway is up-regulated in many human cancers, and agents targeting the mTOR pathway are in various stages of clinical development and application. Expression of pAkt and mTOR was studied by immunohistochemical analysis of 574 surgically resected non-small cell lung cancer (NSCLC) specimens on a tissue microarray. The results were correlated with clinicopathological features. Expression of mTOR showed a strong correlation with the expression of pAkt (p < 0.001) and was significantly associated with female gender, tumor size of ≤3 cm, adenocarcinoma (ADC), non-smoker status, and lower pathological stage. Expression of pAkt was correlated with older age (≥65), ADC, non-smoker status, and lower T stage. Univariate survival analysis revealed that the mTOR- and pAkt-positive group had a significantly longer cancer-specific survival than the mTOR- and pAkt-negative group (p = 0.038 and 0.024, respectively). Coexpression of pAkt and mTOR correlated with better prognosis than either single- or double-negative pAkt and mTOR groups (p = 0.016). However, multivariate analysis proved that mTOR and pAkt expression are not independent prognostic factors for cancer-specific survival. Expression of pAkt and mTOR expression is more significantly associated with ADC than squamous cell carcinoma. Although pAkt/mTOR expression is not an independent prognostic marker, expression of these proteins is associated with better prognosis.
Collapse
Affiliation(s)
- Mee-Hye Oh
- Department of Pathology, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Overexpression of the mammalian target of rapamycin (mTOR) and angioinvasion are poor prognostic factors in early stage NSCLC: a verification study. Lung Cancer 2011; 75:217-22. [PMID: 21802763 DOI: 10.1016/j.lungcan.2011.06.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 06/04/2011] [Accepted: 06/27/2011] [Indexed: 01/04/2023]
Abstract
BACKGROUND A recent study by Dhillon et al. [12], identified both angioinvasion and mTOR as prognostic biomarkers for poor survival in early stage NSCLC. The aim of this study was to verify the above study by examining the angioinvasion and mTOR expression profile in a cohort of early stage NSCLC patients and correlate the results to patient clinico-pathological data and survival. METHODS Angioinvasion was routinely recorded by the pathologist at the initial assessment of the tumor following resection. mTOR was evaluated in 141 early stage (IA-IIB) NSCLC patients (67 - squamous; 60 - adenocarcinoma; 14 - others) using immunohistochemistry (IHC) analysis with an immunohistochemical score (IHS) calculated (% positive cells×staining intensity). Intensity was scored as follows: 0 (negative); 1+ (weak); 2+ (moderate); 3+ (strong). The range of scores was 0-300. Based on the previous study a cut-off score of 30 was used to define positive versus negative patients. The impact of angioinvasion and mTOR expression on prognosis was then evaluated. RESULTS 101 of the 141 tumors studied expressed mTOR. There was no difference in mTOR expression between squamous cell carcinoma and adenocarcinoma. Angioinvasion (p=0.024) and mTOR staining (p=0.048) were significant univariate predictors of poor survival. Both remained significant after multivariate analysis (p=0.037 and p=0.020, respectively). CONCLUSIONS Our findings verify angioinvasion and mTOR expression as new biomarkers for poor outcome in patients with early stage NSCLC. mTOR expressing patients may benefit from novel therapies targeting the mTOR survival pathway.
Collapse
|
40
|
Levy JMM, Thorburn A. Targeting autophagy during cancer therapy to improve clinical outcomes. Pharmacol Ther 2011; 131:130-41. [PMID: 21440002 PMCID: PMC3539744 DOI: 10.1016/j.pharmthera.2011.03.009] [Citation(s) in RCA: 184] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 03/04/2011] [Indexed: 02/06/2023]
Abstract
Autophagy is a catabolic process that turns over long-lived proteins and organelles and contributes to cell and organism survival in times of stress. Current cancer therapies including chemotherapy and radiation are known to induce autophagy within tumor cells. This is therefore an attractive process to target during cancer therapy as there are safe, clinically available drugs known to both inhibit and stimulate autophagy. However, there are conflicting positive and negative effects of autophagy and no current consensus on how to manipulate autophagy to improve clinical outcomes. Careful and rigorous evaluation of autophagy with a focus on how to translate laboratory findings into relevant clinical therapies remains an important aspect of improving clinical outcomes in patients with malignant disease.
Collapse
Affiliation(s)
- Jean M Mulcahy Levy
- Department of Pediatrics University of Colorado 12801 E 17 Ave RC-1 South, Rm 6400D Mail Stop 8303 Aurora, CO, USA, 80045
| | - Andrew Thorburn
- Department of Pharmacology University of Colorado 12801 E 17 Ave RC-1 South, Rm 6400D Mail Stop 8303 Aurora, CO, USA, 80045
| |
Collapse
|
41
|
Klotho predicts good clinical outcome in patients with limited-disease small cell lung cancer who received surgery. Lung Cancer 2011; 74:332-7. [PMID: 21529984 DOI: 10.1016/j.lungcan.2011.03.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 02/01/2011] [Accepted: 03/04/2011] [Indexed: 11/23/2022]
Abstract
BACKGROUND The important role of surgery in early-stage small cell lung cancer (SCLC) has been recognized, and curative surgical resection is recommended. However, the role of adjuvant chemotherapy for stage I SCLC has not yet been evaluated, and novel approaches focusing on the specific genomic characteristics of SCLC may be invaluable for customized therapy. In this study, we focused on the Klotho gene, which is an anti-aging gene known to be a potential tumor suppressor. We investigated whether the expression of Klotho, assessed by immunohistochemistry, can predict survival in patients with resected SCLC. METHODS The medical records of patients diagnosed as having limited-disease (LD) SCLC and treated by surgical resection (n=30) at Tokyo Medical University Hospital were retrospectively reviewed. The expression status of Klotho, and of the ATP-binding cassette (ABC) transporters MRP1, MDR and breast cancer resistant protein (BCRP), which can cause resistance to anticancer drugs, including irinotecan, was assessed by immunohistochemical analysis in resected surgical specimens of patients with early-stage SCLC. RESULTS Of the 30 patients, Klotho expression was seen in the specimens from 18 patients (60.0%), but not in those of the remaining 12 patients (40.0%). The immunostaining for Klotho was mostly localized in the cytoplasm. The expression of Klotho was significantly associated with the overall survival (OS) (ratio 0.088; 95% confidence interval 0.019-0.409; P=0.002). The administration of perioperative chemotherapy had no significant effect in improving the survival, as assessed by the Kaplan-Meier method. However, the patients showing Klotho expression in the resected specimens in p-stage I and II, may have benefited from perioperative chemotherapy. A multivariate analysis revealed no significant association between the expression status of MRP1, MDR or BCRP and the OS. CONCLUSION Expression of Klotho was predictive of a favorable outcome following resection in limited-disease SCLC patients, and the Klotho expression status may serve as a new biomarker for the need of additional therapies to be developed in the future.
Collapse
|
42
|
Liu D, Huang Y, Chen B, Zeng J, Guo N, Zhang S, Liu L, Xu H, Mo X, Li W. Activation of mammalian target of rapamycin pathway confers adverse outcome in nonsmall cell lung carcinoma. Cancer 2011; 117:3763-73. [DOI: 10.1002/cncr.25959] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 12/31/2010] [Accepted: 01/03/2011] [Indexed: 01/11/2023]
|
43
|
Usuda J, Ichinose S, Ishizumi T, Ohtani K, Inoue T, Saji H, Kakihana M, Kajiwara N, Uchida O, Nomura M, Tsutsui H, Ohira T, Ikeda N. Klotho is a novel biomarker for good survival in resected large cell neuroendocrine carcinoma of the lung. Lung Cancer 2010; 72:355-9. [PMID: 21075474 DOI: 10.1016/j.lungcan.2010.10.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 09/08/2010] [Accepted: 10/08/2010] [Indexed: 11/19/2022]
Abstract
BACKGROUND In terms of prognosis, large cell neuroendocrine carcinoma (LCNEC) differs distinctively from other non-small cell lung cancers, with the prognosis of LCNEC being poor, even for early-stage disease. Improvements in survival require a biomarker capable of defining a subset of patients destined to do poorly so that these patients can be targeted for additional therapies, including chemotherapy. In this study, we focused on the Klotho gene, which is an anti-aging gene known to be a potential tumor suppressor. We investigated whether the immunohistochemical expression of Klotho can predict survival patients with resected LCNEC. METHODS The histological characteristics of patients receiving an initial diagnosis of LCNEC (n=30) at Tokyo Medical University Hospital were retrospectively reviewed, and multiple variables including stage, lymphangioinvasion, lymph node status and the expression of Klotho as identified using an immunohistochemical analysis, were assessed. RESULTS Immunostaining for Klotho was mostly cytoplasmic, and Klotho expression was seen in 10 patients (33.3%) but not in 20 patients (66.7%). The expression of Klotho was significantly associated with a good outcome of resected patients with LCNEC and Klotho(-) was associated with increased LCNEC risk by multivariate analysis (hazard ratio 4.92, 95% confidence interval 1.04-23.24, p=0.044). Neither lymph node status nor lymphangioinvasion were significantly associated with a poor survival. However, among patients without lymph node metastasis or angioinvasion, the survival benefit of Klotho expression in the primary tumor was significantly higher, compared with that of patients without Klotho expression. CONCLUSION Klotho staining provides a new biomarker for a good outcome in patients with LCNEC, especially among patients without lymph node metastasis or lymphangioinvasion.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Large Cell/diagnosis
- Carcinoma, Large Cell/mortality
- Carcinoma, Large Cell/pathology
- Carcinoma, Large Cell/physiopathology
- Carcinoma, Large Cell/surgery
- Carcinoma, Neuroendocrine/diagnosis
- Carcinoma, Neuroendocrine/mortality
- Carcinoma, Neuroendocrine/pathology
- Carcinoma, Neuroendocrine/physiopathology
- Carcinoma, Neuroendocrine/surgery
- Female
- Glucuronidase/genetics
- Glucuronidase/metabolism
- Humans
- Immunohistochemistry
- Klotho Proteins
- Lung Neoplasms/diagnosis
- Lung Neoplasms/mortality
- Lung Neoplasms/pathology
- Lung Neoplasms/physiopathology
- Lung Neoplasms/surgery
- Lymphatic Metastasis
- Male
- Middle Aged
- Neoplasm Staging
- Predictive Value of Tests
- Prognosis
- Retrospective Studies
- Survival Analysis
Collapse
Affiliation(s)
- Jitsuo Usuda
- Division of Thoracic Surgery 1, Department of Surgery 1, Tokyo Medical University, 6-7-1, Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kaira K, Yamamoto N. Prognostic and predictive factors in resected non-small-cell lung cancer. ACTA ACUST UNITED AC 2010; 4:373-81. [DOI: 10.1517/17530059.2010.506214] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|