1
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [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
|
2
|
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: 15] [Impact Index Per Article: 15.0] [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
|
3
|
Ji L, Moghal N, Zou X, Fang Y, Hu S, Wang Y, Tsao MS. The NRF2 antagonist ML385 inhibits PI3K-mTOR signaling and growth of lung squamous cell carcinoma cells. Cancer Med 2023; 12:5688-5702. [PMID: 36305267 PMCID: PMC10028163 DOI: 10.1002/cam4.5311] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/11/2022] [Accepted: 09/20/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Lung squamous cell carcinoma (LUSC) currently has limited therapeutic options because of the relatively few validated targets and the lack of clinical drugs for some of these targets. Although NRF2/NFE2L2 pathway activation commonly occurs in LUSC, NRF2 has predominantly been studied in other cancer models. Here, we investigated the function of NRF2 in LUSC, including in organoid models, and we explored the activity of a small molecule NRF2 inhibitor ML385, which has not previously been investigated in LUSC. METHODS We first explored the role of NRF2 signaling in LUSC cancer cell line and organoid proliferation through NRF2 knockdown or ML385 treatment, both in vivo and in vitro. Next, we performed Western blot and immunofluorescence assays to determine the effect of NRF2 inhibition on PI3K-mTOR signaling. Finally, we used cell viability and clonogenic assays to explore whether ML385 could sensitize LUSC cancer cells to PI3K inhibitors. RESULTS We find that downregulation of NRF2 signaling inhibited proliferation of LUSC cancer cell lines and organoids, both in vivo and in vitro. We also demonstrate that inhibition of NRF2 reduces PI3K-mTOR signaling, with two potential mechanisms being involved. Although NRF2 promotes AKT phosphorylation, it also acts downstream of AKT to increase RagD protein expression and recruitment of mTOR to lysosomes after amino acid stimulation. We also find that ML385 potentiates LUSC growth inhibition by a pan-PI3K inhibitor, which correlates with stronger inhibition of PI3K-mTOR signaling. CONCLUSIONS Our data provide additional support for NRF2 promoting LUSC growth through PI3K-mTOR activation and support development of NRF2 inhibitors for the treatment of LUSC.
Collapse
Affiliation(s)
- Lili Ji
- Department of Pathology, Key Laboratory of Microenvironment and Translational Cancer Research, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Nadeem Moghal
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Xinru Zou
- Department of Pathology, Key Laboratory of Microenvironment and Translational Cancer Research, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Yixuan Fang
- Department of Pathology, Key Laboratory of Microenvironment and Translational Cancer Research, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Shuning Hu
- Department of Pathology, Key Laboratory of Microenvironment and Translational Cancer Research, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Yuhui Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ming Sound Tsao
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Lan J, Zhu Y, Rao J, Liu L, Gong A, Feng F, Chen B, Huang J, Zhang Y, Chu L, Zhong H, Li L, Yan J, Li W, Xue C. MTOR gene polymorphism may be associated with microscopic polyangiitis susceptibility in a Guangxi population of China. Gene X 2023; 854:147101. [PMID: 36496178 DOI: 10.1016/j.gene.2022.147101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/18/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Microscopic polyangiitis (MPA) onset is affected by genetic predisposition. Autophagy plays a certain role in antineutrophil cytoplasmic antibody-associated vasculitis developing. A key factor in autophagy regulating, the genetic polymorphism of MTOR gene is essential. The objective was to explore the associations between MTOR gene polymorphism and MPA susceptibility in a Guangxi population of China. METHODS A sum of 208 MPA cases and 209 healthy volunteers from Guangxi in this case-control study, four important single nucleotide polymorphism (SNP) loci of MTOR gene including rs3806317, rs1064261, rs1883965 and rs2295080 were examined. Multiplex polymerase chain reaction combined with high-throughput sequencing was performed. Subgroup analysis was evaluated by gender and ethnicity. Linkage disequilibrium and haplotype analysis were tested. Multi-SNPs interaction among mTOR signaling pathway was assessed. RESULTS For rs2295080, homozygous mutant GG genotype was associated with a decreased susceptibility of MPA in recessive model (OR = 0.38, 95%CI: 0.14-1.00, p = 0.040), particularly in the subgroup of female (OR = 0.16, 95%CI: 0.03-0.74, p = 0.006) and Han population (OR = 0.32, 95%CI: 0.10-1.00, p = 0.034). Individual carrying G allele was linked with decreasing MPA susceptibility in Han population of Guangxi (OR = 0.65, 95%CI: 0.44-0.97, p = 0.036). In haplotype analysis, the haplotype AAT was correlated with increasing susceptibility of MPA (OR = 1.347, 95%CI: 1.004-1.807, p = 0.046). Moreover, in the multi-SNPs interaction analysis, the six-locus model was identified as the best interaction model (p < 0.05). CONCLUSION These findings suggest that rs2295080 polymorphism of MTOR gene may be associated with MPA susceptibility in a Guangxi population of China and G allele might be an important protective factor.
Collapse
Affiliation(s)
- Jingjing Lan
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Yan Zhu
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China; The First Affiliated Hospital, Department of Nephrology, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
| | - Jinlan Rao
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Liu Liu
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Aimei Gong
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China; Department of Nephrology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, P.R. China
| | - Fei Feng
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Bingfang Chen
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Junxia Huang
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Yurong Zhang
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Liepeng Chu
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Huan Zhong
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Lizhen Li
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China; Department of Nephrology, Hunan Research Institute of Geriatrics, The Peoples Hospital of Hunan Province, Changsha, Hunan, P.R. China
| | - Jinlian Yan
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Wei Li
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China.
| | - Chao Xue
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China.
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Abstract
ABSTRACT The phosphosphatidylinositol-3-kinase (PI3K) signaling pathway is one of the most important intracellular signal transduction pathways affecting cell functions, such as apoptosis, translation, metabolism, and angiogenesis. Lung cancer is a malignant tumor with the highest morbidity and mortality rates in the world. It can be divided into two groups, non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). NSCLC accounts for >85% of all lung cancers. There are currently many clinical treatment options for NSCLC; however, traditional methods such as surgery, chemotherapy, and radiotherapy have not been able to provide patients with good survival benefits. The emergence of molecular target therapy has improved the survival and prognosis of patients with NSCLC. In recent years, there have been an increasing number of studies on NSCLC and PI3K signaling pathways. Inhibitors of various parts of the PI3K pathway have appeared in various phases of clinical trials with NSCLC as an indication. This article focuses on the role of the PI3K signaling pathway in the occurrence and development of NSCLC and summarizes the current clinical research progress and possible development strategies.
Collapse
|
8
|
Targeting sphingosine kinase 1/2 by a novel dual inhibitor SKI-349 suppresses non-small cell lung cancer cell growth. Cell Death Dis 2022; 13:602. [PMID: 35831279 PMCID: PMC9279331 DOI: 10.1038/s41419-022-05049-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/16/2022] [Accepted: 06/27/2022] [Indexed: 01/21/2023]
Abstract
Sphingosine kinase 1 (SphK1) and sphingosine kinase (SphK2) are both important therapeutic targets of non-small cell lung cancer (NSCLC). SKI-349 is a novel, highly efficient and small molecular SphK1/2 dual inhibitor. Here in primary human NSCLC cells and immortalized cell lines, SKI-349 potently inhibited cell proliferation, cell cycle progression, migration and viability. The dual inhibitor induced mitochondrial depolarization and apoptosis activation in NSCLC cells, but it was non-cytotoxic to human lung epithelial cells. SKI-349 inhibited SphK activity and induced ceramide accumulation in primary NSCLC cells, without affecting SphK1/2 expression. SKI-349-induced NSCLC cell death was attenuated by sphingosine-1-phosphate and by the SphK activator K6PC-5, but was potentiated by the short-chain ceramide C6. Moreover, SKI-349 induced Akt-mTOR inactivation, JNK activation, and oxidative injury in primary NSCLC cells. In addition, SKI-349 decreased bromodomain-containing protein 4 (BRD4) expression and downregulated BRD4-dependent genes (Myc, cyclin D1 and Klf4) in primary NSCLC cells. At last, SKI-349 (10 mg/kg) administration inhibited NSCLC xenograft growth in nude mice. Akt-mTOR inhibition, JNK activation, oxidative injury and BRD4 downregulation were detected in SKI-349-treated NSCLC xenograft tissues. Taken together, targeting SphK1/2 by SKI-349 potently inhibits NSCLC cell growth in vitro and in vivo.
Collapse
|
9
|
Zakharova VV, Magnitov MD, Del Maestro L, Ulianov SV, Glentis A, Uyanik B, Williart A, Karpukhina A, Demidov O, Joliot V, Vassetzky Y, Mège RM, Piel M, Razin S, Ait-Si-Ali S. SETDB1 fuels the lung cancer phenotype by modulating epigenome, 3D genome organization and chromatin mechanical properties. Nucleic Acids Res 2022; 50:4389-4413. [PMID: 35474385 PMCID: PMC9071401 DOI: 10.1093/nar/gkac234] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/15/2022] [Accepted: 04/21/2022] [Indexed: 12/24/2022] Open
Abstract
Imbalance in the finely orchestrated system of chromatin-modifying enzymes is a hallmark of many pathologies such as cancers, since causing the affection of the epigenome and transcriptional reprogramming. Here, we demonstrate that a loss-of-function mutation (LOF) of the major histone lysine methyltransferase SETDB1 possessing oncogenic activity in lung cancer cells leads to broad changes in the overall architecture and mechanical properties of the nucleus through genome-wide redistribution of heterochromatin, which perturbs chromatin spatial compartmentalization. Together with the enforced activation of the epithelial expression program, cytoskeleton remodeling, reduced proliferation rate and restricted cellular migration, this leads to the reversed oncogenic potential of lung adenocarcinoma cells. These results emphasize an essential role of chromatin architecture in the determination of oncogenic programs and illustrate a relationship between gene expression, epigenome, 3D genome and nuclear mechanics.
Collapse
Affiliation(s)
- Vlada V Zakharova
- Epigenetics and Cell Fate (EDC) department, UMR7216, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Mikhail D Magnitov
- Institute of Gene Biology, Russian Academy of Science, Moscow 119334, Russia
| | - Laurence Del Maestro
- Epigenetics and Cell Fate (EDC) department, UMR7216, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Sergey V Ulianov
- Institute of Gene Biology, Russian Academy of Science, Moscow 119334, Russia,Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow 119991, Russia
| | - Alexandros Glentis
- Institute Jacques Monod, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Burhan Uyanik
- INSERM UMR1231, LipSTIC, University of Burgundy Franche-Comté F-21000, Dijon, France
| | - Alice Williart
- Institut Curie and Institut Pierre Gilles de Gennes, PSL Research University, CNRS, UMR 144, 75248 Paris, France
| | - Anna Karpukhina
- UMR9018, CNRS, Université Paris-Sud Paris-Saclay, Institut Gustave Roussy; 94805 Villejuif, France,Koltzov Institute of Developmental Biology, 119334 Moscow, Russia
| | - Oleg Demidov
- INSERM UMR1231, LipSTIC, University of Burgundy Franche-Comté F-21000, Dijon, France,Institute of Cytology, RAS, 194064 St. Petersburg, Russia,NTU Sirius, 354340 Sochi, Russia
| | - Veronique Joliot
- Epigenetics and Cell Fate (EDC) department, UMR7216, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Yegor S Vassetzky
- UMR9018, CNRS, Université Paris-Sud Paris-Saclay, Institut Gustave Roussy; 94805 Villejuif, France,Koltzov Institute of Developmental Biology, 119334 Moscow, Russia
| | - René-Marc Mège
- Institute Jacques Monod, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Matthieu Piel
- Institut Curie and Institut Pierre Gilles de Gennes, PSL Research University, CNRS, UMR 144, 75248 Paris, France
| | - Sergey V Razin
- Correspondence may also be addressed to Sergey V. Razin. Tel: +7 499 135 3092;
| | | |
Collapse
|
10
|
Kraaijvanger R, Seldenrijk K, Beijer E, Damen J, Wilson JL, Weichhart T, Grutters JC, Veltkamp M. Activation of Downstream mTORC1 Target Ribosomal Protein S6 Kinase (S6K) Can Be Found in a Subgroup of Dutch Patients with Granulomatous Pulmonary Disease. Cells 2021; 10:3545. [PMID: 34944053 PMCID: PMC8700352 DOI: 10.3390/cells10123545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 11/16/2022] Open
Abstract
Mechanistic target of rapamycin complex 1 (mTORC1) has been linked to different diseases. The mTORC1 signaling pathway is suggested to play a role in the granuloma formation of sarcoidosis. Recent studies demonstrated conflicting data on mTORC1 activation in patients with sarcoidosis by measuring activation of its downstream target S6 kinase (S6K) with either 33% or 100% of patients. Therefore, the aim of our study was to reevaluate the percentage of S6K activation in sarcoidosis patients in a Dutch cohort. To investigate whether this activation is specific for sarcoid granulomas, we also included Dutch patients with other granulomatous diseases of the lung. The activation of the S6K signaling pathway was evaluated by immunohistochemical staining of its downstream effector phospho-S6 in tissue sections. Active S6K signaling was detected in 32 (43%) of the sarcoidosis patients. Twelve (31%) of the patients with another granulomatous disorder also showed activated S6K signaling, demonstrating that the mTORC1 pathway may be activated in a range for different granulomatous diseases (p = 0.628). Activation of S6K can only be found in a subgroup of patients with sarcoidosis, as well as in patients with other granulomatous pulmonary diseases, such as hypersensitivity pneumonitis or vasculitis. No association between different clinical phenotypes and S6K activation can be found in sarcoidosis.
Collapse
Affiliation(s)
- Raisa Kraaijvanger
- Interstitial Lung Diseases Centre of Excellence, Department of Pulmonology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands; (R.K.); (E.B.); (J.C.G.)
| | - Kees Seldenrijk
- Interstitial Lung Diseases Centre of Excellence, Pathology DNA, Department of Pathology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands;
| | - Els Beijer
- Interstitial Lung Diseases Centre of Excellence, Department of Pulmonology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands; (R.K.); (E.B.); (J.C.G.)
| | - Jan Damen
- Pathology DNA, Department of Pathology, Jeroen Bosch Hospital, 5223 GZ ’s-Hertogenbosch, The Netherlands;
| | - Jayne Louise Wilson
- Center for Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, 1090 Vienna, Austria; (J.L.W.); (T.W.)
| | - Thomas Weichhart
- Center for Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, 1090 Vienna, Austria; (J.L.W.); (T.W.)
| | - Jan C. Grutters
- Interstitial Lung Diseases Centre of Excellence, Department of Pulmonology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands; (R.K.); (E.B.); (J.C.G.)
- Division of Hearth and Lungs, University Medical Centre, 3584 CX Utrecht, The Netherlands
| | - Marcel Veltkamp
- Interstitial Lung Diseases Centre of Excellence, Department of Pulmonology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands; (R.K.); (E.B.); (J.C.G.)
- Division of Hearth and Lungs, University Medical Centre, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
11
|
Autophagic Activation and Decrease of Plasma Membrane Cholesterol Contribute to Anticancer Activities in Non-Small Cell Lung Cancer. Molecules 2021; 26:molecules26195967. [PMID: 34641511 PMCID: PMC8512437 DOI: 10.3390/molecules26195967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 12/25/2022] Open
Abstract
Non-small cell lung cancer (NSCLC), an aggressive subtype of pulmonary carcinomas with high mortality, accounts for 85% of all lung cancers. Drug resistance and high recurrence rates impede the chemotherapeutic effect, making it urgent to develop new anti-NSCLC agents. Recently, we have demonstrated that para-toluenesulfonamide is a potential anti-tumor agent in human castration-resistant prostate cancer (CRPC) through inhibition of Akt/mTOR/p70S6 kinase pathway and lipid raft disruption. In the current study, we further addressed the critical role of cholesterol-enriched membrane microdomain and autophagic activation to para-toluenesulfonamide action in killing NSCLC. Similar in CRPC, para-toluenesulfonamide inhibited the Akt/mTOR/p70S6K pathway in NSCLC cell lines NCI-H460 and A549, leading to G1 arrest of the cell cycle and apoptosis. Para-toluenesulfonamide significantly decreased the cholesterol levels of plasma membrane. External cholesterol supplement rescued para-toluenesulfonamide-mediated effects. Para-toluenesulfonamide induced a profound increase of LC3-II protein expression and a significant decrease of p62 expression. Double staining of lysosomes and cellular cholesterol showed para-toluenesulfonamide-induced lysosomal transportation of cholesterol, which was validated using flow cytometric analysis of lysosome staining. Moreover, autophagy inhibitors could blunt para-toluenesulfonamide-induced effect, indicating autophagy induction. In conclusion, the data suggest that para-toluenesulfonamide is an effective anticancer agent against NSCLC through G1 checkpoint arrest and apoptotic cell death. The disturbance of membrane cholesterol levels and autophagic activation may play a crucial role to para-toluenesulfonamide action.
Collapse
|
12
|
Pizzini A, Bacher H, Aichner M, Franchi A, Watzinger K, Tancevski I, Sonnweber T, Mosheimer-Feistritzer B, Duftner C, Zelger B, Pallua J, Sprung S, Weichhart T, Zelger B, Weiss G, Löffler-Ragg J. High expression of mTOR signaling in granulomatous lesions is not predictive for the clinical course of sarcoidosis. Respir Med 2021; 177:106294. [PMID: 33485108 DOI: 10.1016/j.rmed.2020.106294] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 08/31/2020] [Accepted: 12/26/2020] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Sarcoidosis is a systemic granulomatous disease with a variable clinical presentation and disease course. There is still no reliable biomarker available, which assists in the diagnosis or prediction of the clinical course. According to a murine model, the expression level of the metabolic checkpoint kinase mechanistic target of Rapamycin complex 1 (mTORC1) in granulomas of sarcoidosis patients may be used as a clinical biomarker. MATERIAL AND METHODS This is a retrospective analysis of 58 patients with histologically confirmed sarcoidosis. Immunohistochemical staining of granulomas from tissue samples was evaluated for the expression of activated mTORC1 signaling, including phosphorylated mTOR, its downstream effectors S6K1, 4EBP1 and the proliferation marker Ki-67. Patients were categorized according to different clinical phenotypes, serum biomarkers, and immunomodulatory therapy. RESULTS All patients showed activated mTORC1 signaling in granulomas, which correlated with its downstream effectors S6K1 and 4EBP1 but was not related to Ki-67 expression. The mTORC1 activity revealed an association neither to disease severity nor the necessity of treatment; however, p-mTOR inversely correlated with cumulative corticosteroid dosage. CONCLUSION Our data confirm activation of the mTORC1 pathway in sarcoidosis, supporting the hypothesis that mTOR is a significant driver in granuloma formation. However, we could not find a relationship between the degree of mTOR activation and disease severity or the need for therapy.
Collapse
Affiliation(s)
- Alex Pizzini
- Medical University of Innsbruck, Department of Internal Medicine II, Anichstraße 35, 6020, Innsbruck, Austria
| | - Hannes Bacher
- Medical University of Innsbruck, Department of Internal Medicine II, Anichstraße 35, 6020, Innsbruck, Austria
| | - Magdalena Aichner
- Medical University of Innsbruck, Department of Internal Medicine II, Anichstraße 35, 6020, Innsbruck, Austria
| | - Alexander Franchi
- Medical University of Innsbruck, Department of Internal Medicine II, Anichstraße 35, 6020, Innsbruck, Austria
| | - Kathrin Watzinger
- Medical University of Innsbruck, Department of Internal Medicine II, Anichstraße 35, 6020, Innsbruck, Austria
| | - Ivan Tancevski
- Medical University of Innsbruck, Department of Internal Medicine II, Anichstraße 35, 6020, Innsbruck, Austria
| | - Thomas Sonnweber
- Medical University of Innsbruck, Department of Internal Medicine II, Anichstraße 35, 6020, Innsbruck, Austria
| | | | - Christina Duftner
- Medical University of Innsbruck, Department of Internal Medicine II, Anichstraße 35, 6020, Innsbruck, Austria
| | - Bettina Zelger
- Medical University of Innsbruck, Department of Pathology, General Pathology Division, Anichstraße 35, 6020, Innsbruck, Austria
| | - Johannes Pallua
- Medical University of Innsbruck, Department of Pathology, General Pathology Division, Anichstraße 35, 6020, Innsbruck, Austria
| | - Susanne Sprung
- Medical University of Innsbruck, Department of Pathology, General Pathology Division, Anichstraße 35, 6020, Innsbruck, Austria
| | - Thomas Weichhart
- Medical University of Vienna, Institute of Medical Genetics, Währinger Straße 10, 1090, Vienna, Austria
| | - Bernhard Zelger
- Medical University of Innsbruck, Department of Dermatology and Venereology, Anichstraße 35, 6020, Innsbruck, Austria
| | - Günter Weiss
- Medical University of Innsbruck, Department of Internal Medicine II, Anichstraße 35, 6020, Innsbruck, Austria.
| | - Judith Löffler-Ragg
- Medical University of Innsbruck, Department of Internal Medicine II, Anichstraße 35, 6020, Innsbruck, Austria.
| |
Collapse
|
13
|
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: 0.8] [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
|
14
|
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: 281] [Impact Index Per Article: 70.3] [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
|
15
|
Ding C, Zhou X, Xu C, Chen J, Ju S, Chen T, Liang Z, Cui Z, Li C, Zhao J. Circulating tumor cell levels and carcinoembryonic antigen: An improved diagnostic method for lung adenocarcinoma. Thorac Cancer 2018; 9:1413-1420. [PMID: 30178907 PMCID: PMC6209798 DOI: 10.1111/1759-7714.12851] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 07/27/2018] [Accepted: 07/27/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The aim of this study was to determine a correlation between benign and malignant lung solitary pulmonary nodules (SPN), and analyze the association between circulating tumor cell (CTC) levels and different subtypes of lung adenocarcinoma. METHODS A total of 200 patients (80 with SPNs and 120 diagnosed with lung cancer) were included in the study. The CTC levels were quantified by identifying the folate receptor on the surface of tumor cells; clinical tumor specific markers were detected by biochemical immunization. The content of peripheral blood CTCs in benign and malignant lung SPN patients was detected and the differences in preoperative CTC levels in different pathological subtypes were analyzed. Based on the collected data, receiver operating characteristic curves were calculated and the rate of lung cancer was predicted. RESULTS The peripheral blood CTC levels in patients with malignant lung SPNs were higher than in patients with benign SPNs. The maximum nodule diameter, carcinoembryonic antigen, and CTC levels were independent risk factors for malignant lung SPNs. The peripheral blood CTC levels in patients with stage III-IV lung adenocarcinoma were higher than in stage I-II patients. The peripheral blood CTC levels in patients with microinvasive and invasive adenocarcinoma were higher than in adenocarcinoma in situ patients. The CTC levels in the peripheral blood of patients with maximum tumor diameter > 2 cm were higher than in patients with tumors < 2 cm. CONCLUSION The detection of CTCs can be used as a biomarker for screening SPNs and diagnosing early-stage lung cancer. Using the combination of CTC levels and CEA significantly improves the efficacy of lung adenocarcinoma diagnosis.
Collapse
Affiliation(s)
- Cheng Ding
- Department of Thoracic SurgeryThe First Affiliated Hospital of Soochow University, Medical College of Soochow UniversitySuzhouChina
| | - Xiaofei Zhou
- Department of RadiologyThe First Affiliated Hospital of Soochow University, Medical College of Soochow UniversitySuzhouChina
| | - Chun Xu
- Department of Thoracic SurgeryThe First Affiliated Hospital of Soochow University, Medical College of Soochow UniversitySuzhouChina
| | - Jun Chen
- Department of Thoracic SurgeryThe First Affiliated Hospital of Soochow University, Medical College of Soochow UniversitySuzhouChina
| | - Shen Ju
- Department of Thoracic SurgeryThe First Affiliated Hospital of Soochow University, Medical College of Soochow UniversitySuzhouChina
| | - Tengfei Chen
- Department of Thoracic SurgeryThe First Affiliated Hospital of Soochow University, Medical College of Soochow UniversitySuzhouChina
| | - Zhipan Liang
- Department of Thoracic SurgeryThe First Affiliated Hospital of Soochow University, Medical College of Soochow UniversitySuzhouChina
| | - Zihan Cui
- Department of Thoracic SurgeryThe First Affiliated Hospital of Soochow University, Medical College of Soochow UniversitySuzhouChina
| | - Chang Li
- Department of Thoracic SurgeryThe First Affiliated Hospital of Soochow University, Medical College of Soochow UniversitySuzhouChina
| | - Jun Zhao
- Department of Thoracic SurgeryThe First Affiliated Hospital of Soochow University, Medical College of Soochow UniversitySuzhouChina
| |
Collapse
|
16
|
Heavey S, Cuffe S, Finn S, Young V, Ryan R, Nicholson S, Leonard N, McVeigh N, Barr M, O'Byrne K, Gately K. In pursuit of synergy: An investigation of the PI3K/mTOR/MEK co-targeted inhibition strategy in NSCLC. Oncotarget 2018; 7:79526-79543. [PMID: 27765909 PMCID: PMC5346733 DOI: 10.18632/oncotarget.12755] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 09/12/2016] [Indexed: 12/20/2022] Open
Abstract
Clinical PI3K inhibition has been somewhat disappointing, due to both inadequate patient stratification and compensatory cell signalling through bypass mechanisms. As such, investigation of PI3K-MEK co-targeted inhibition has been recommended. With high mortality rates and a clear need for new therapeutic intervention strategies, non-small cell lung cancer (NSCLC) is an important setting to investigate the effectiveness of this approach. Here, 174 NSCLC tumours were screened for 150 mutations by Fluidigm technology, with 15 patients being profiled for phosphoprotein expression. The effects of GDC-0941 (a pan PI3K inhibitor), GDC-0980 (a dual PI3K/mTOR inhibitor) and GDC-0973 (a MEK inhibitor) alone and in combination were assessed in 3 NSCLC cell lines. PIK3CA was mutated in 5.17% of NSCLC patients. GDC-0941 and GDC-0980 treatment induced anti-proliferative and pro-apoptotic responses across all NSCLC cell lines, while GDC-0973 treatment induced only anti-proliferative responses. GDC-0980 and GDC-0973 combined treatment led to significant increases in apoptosis and synergistic reductions in proliferation across the panel of cell lines. This study found that the PI3K/MEK co-targeted inhibition strategy is synergistic in all 3 molecular subtypes of NSCLC investigated. Consequently, we would advocate clinical trials for NSCLC patients combining GDC-0980 and GDC-0973, each of which are separately under clinical investigation currently.
Collapse
Affiliation(s)
- Susan Heavey
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| | - Sinead Cuffe
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| | - Stephen Finn
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| | - Vincent Young
- Department of Cardiothoracic Surgery, St. James's Hospital, Dublin, Ireland
| | - Ronan Ryan
- Department of Cardiothoracic Surgery, St. James's Hospital, Dublin, Ireland
| | | | - Niamh Leonard
- Department of Histopathology, St James Hospital, Dublin, Ireland
| | - Niall McVeigh
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| | - Martin Barr
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| | - Kenneth O'Byrne
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation at the Translational Research Institute (TRI), Queensland University of Technology, Brisbane, Australia
| | - Kathy Gately
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
17
|
Guglielmelli T, Giugliano E, Brunetto V, Rapa I, Cappia S, Giorcelli J, Rrodhe S, Papotti M, Saglio G. mTOR pathway activation in multiple myeloma cell lines and primary tumour cells: pomalidomide enhances cytoplasmic-nuclear shuttling of mTOR protein. Oncoscience 2015; 2:382-94. [PMID: 26097872 PMCID: PMC4468324 DOI: 10.18632/oncoscience.148] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/16/2015] [Indexed: 01/23/2023] Open
Abstract
mTOR is a protein kinase that plays a central role in regulating critical cellular processes. We evaluated the activation and cellular localization of the mTOR pathway in multiple myeloma (MM) and analyzed the role of pomalidomide in regulating mTOR. By immunohistochemistry cytoplasmic p-mTOR stained positive in 57 out 101 (57.6%) cases with a nuclear p-mTOR localization in 14 out 101 samples (13.8%). In the 70 MM samples analyzed for the entire pathway, p-mTOR expression significantly correlated with p-AKT, p-P70S6K, and p-4E-BP1 suggesting that the AKT/mTOR pathway is activated in a subset of MM patients. Immunofluorescence assays demonstrated that mTOR protein is distributed throughout the cytoplasm and the nucleus at baseline in MM cell lines and in plasma cells of 13 MM patients and that pomalidomide facilitated the shift of the mTOR protein in the nucleus. By western blotting, treatment with pomalidomide increased nuclear mTOR and p-mTOR expression levels in the nucleus with a concomitant decrease of the cytoplasmic fractions while does not seem to affect significantly AKT phosphorylation status. In MM cells the anti-myeloma activity of pomalidomide may be mediated by the regulation of the mTOR pathway.
Collapse
Affiliation(s)
- Tommasina Guglielmelli
- Department of Clinical and Biological Sciences, University of Turin and S Luigi Hospital, Orbassano, Turin, Italy
| | - Emilia Giugliano
- Department of Clinical and Biological Sciences, University of Turin and S Luigi Hospital, Orbassano, Turin, Italy
| | - Vanessa Brunetto
- Department of Clinical and Biological Sciences, University of Turin and S Luigi Hospital, Orbassano, Turin, Italy
| | - Ida Rapa
- Department of Oncology, University of Turin and S Luigi Hospital, Orbassano, Turin, Italy
| | - Susanna Cappia
- Department of Oncology, University of Turin and S Luigi Hospital, Orbassano, Turin, Italy
| | - Jessica Giorcelli
- Department of Oncology, University of Turin and S Luigi Hospital, Orbassano, Turin, Italy
| | - Sokol Rrodhe
- Department of Clinical and Biological Sciences, University of Turin and S Luigi Hospital, Orbassano, Turin, Italy
| | - Mauro Papotti
- Department of Oncology, University of Turin and S Luigi Hospital, Orbassano, Turin, Italy
| | - Giuseppe Saglio
- Department of Clinical and Biological Sciences, University of Turin and S Luigi Hospital, Orbassano, Turin, Italy
| |
Collapse
|
18
|
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: 2.0] [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
|
19
|
Rodriguez-Teja M, Gronau JH, Breit C, Zhang YZ, Minamidate A, Caley MP, McCarthy A, Cox TR, Erler JT, Gaughan L, Darby S, Robson C, Mauri F, Waxman J, Sturge J. AGE-modified basement membrane cooperates with Endo180 to promote epithelial cell invasiveness and decrease prostate cancer survival. J Pathol 2014; 235:581-92. [PMID: 25408555 DOI: 10.1002/path.4485] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 11/05/2014] [Accepted: 11/08/2014] [Indexed: 11/10/2022]
Abstract
Biomechanical strain imposed by age-related thickening of the basal lamina and augmented tissue stiffness in the prostate gland coincides with increased cancer risk. Here we hypothesized that the structural alterations in the basal lamina associated with age can induce mechanotransduction pathways in prostate epithelial cells (PECs) to promote invasiveness and cancer progression. To demonstrate this, we developed a 3D model of PEC acini in which thickening and stiffening of basal lamina matrix was induced by advanced glycation end-product (AGE)-dependent non-enzymatic crosslinking of its major components, collagen IV and laminin. We used this model to demonstrate that antibody targeted blockade of CTLD2, the second of eight C-type lectin-like domains in Endo180 (CD280, CLEC13E, KIAA0709, MRC2, TEM9, uPARAP) that can recognize glycosylated collagens, reversed actinomyosin-based contractility [myosin-light chain-2 (MLC2) phosphorylation], loss of cell polarity, loss of cell-cell junctions, luminal infiltration and basal invasion induced by AGE-modified basal lamina matrix in PEC acini. Our in vitro results were concordant with luminal occlusion of acini in the prostate glands of adult Endo180(Δ) (Ex2-6/) (Δ) (Ex2-6) mice, with constitutively exposed CTLD2 and decreased survival of men with early (non-invasive) prostate cancer with high epithelial Endo180 expression and levels of AGE. These findings indicate that AGE-dependent modification of the basal lamina induces invasive behaviour in non-transformed PECs via a molecular mechanism linked to cancer progression. This study provides a rationale for targeting CTLD2 in Endo180 in prostate cancer and other pathologies in which increased basal lamina thickness and tissue stiffness are driving factors. © 2014 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Mercedes Rodriguez-Teja
- Department of Surgery and Cancer, Imperial College London, UK; Departamento de Genética, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Rodriguez-Teja M, Gronau JH, Minamidate A, Darby S, Gaughan L, Robson C, Mauri F, Waxman J, Sturge J. Survival Outcome and EMT Suppression Mediated by a Lectin Domain Interaction of Endo180 and CD147. Mol Cancer Res 2014; 13:538-47. [PMID: 25381222 DOI: 10.1158/1541-7786.mcr-14-0344-t] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Epithelial cell-cell contacts maintain normal glandular tissue homeostasis, and their breakage can trigger epithelial-to-mesenchymal transition (EMT), a fundamental step in the development of metastatic cancer. Despite the ability of C-type lectin domains (CTLD) to modulate cell-cell adhesion, it is not known if they modulate epithelial adhesion in EMT and tumor progression. Here, the multi-CTLD mannose receptor, Endo180 (MRC2/uPARAP), was shown using the Kaplan-Meier analysis to be predictive of survival outcome in men with early prostate cancer. A proteomic screen of novel interaction partners with the fourth CTLD (CTLD4) in Endo180 revealed that its complex with CD147 is indispensable for the stability of three-dimensional acini formed by nontransformed prostate epithelial cells (PEC). Mechanistic study using knockdown of Endo180 or CD147, and treatment with an Endo180 mAb targeting CTLD4 (clone 39.10), or a dominant-negative GST-CTLD4 chimeric protein, induced scattering of PECs associated with internalization of Endo180 into endosomes, loss of E-cadherin (CDH1/ECAD), and unzipping of cell-cell junctions. These findings are the first to demonstrate that a CTLD acts as a suppressor and regulatory switch for EMT; thus, positing that stabilization of Endo180-CD147 complex is a viable therapeutic strategy to improve rates of prostate cancer survival. IMPLICATIONS This study identifies the interaction between CTLD4 in Endo180 and CD147 as an EMT suppressor and indicates that stabilization of this molecular complex improves prostate cancer survival rates.
Collapse
Affiliation(s)
- Mercedes Rodriguez-Teja
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom. Departamento de Genética, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Julian H Gronau
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Ai Minamidate
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Steven Darby
- Northern Institute for Cancer Research, Newcastle University Medical School, Newcastle upon Tyne, United Kingdom
| | - Luke Gaughan
- Northern Institute for Cancer Research, Newcastle University Medical School, Newcastle upon Tyne, United Kingdom
| | - Craig Robson
- Northern Institute for Cancer Research, Newcastle University Medical School, Newcastle upon Tyne, United Kingdom
| | - Francesco Mauri
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Jonathan Waxman
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Justin Sturge
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom. School of Biological, Biomedical and Environmental Sciences, University of Hull, Hull, United Kingdom.
| |
Collapse
|
21
|
Zhang Y, Lu H, Xu G. [Effect of PI3K/AKT pathway on cisplatin resistance in non-small cell lung cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2014; 17:635-42. [PMID: 25130971 PMCID: PMC6000368 DOI: 10.3779/j.issn.1009-3419.2014.08.09] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Accumulating evidences indicate that aberrant activation of PI3K/AKT pathway in non-small cell lung cancer plays a vital role in tumor cell proliferation,apoptosis, and survival including drug resistance. Cisplatin as first-line chemotherapy are in widespread clinical use in patients with non-small cell lung cancer, however, the development of cisplatin resistance significantly impedes its clinic efficacy. Cisplatin resistance is a complicated process that various mechanisms participating in to interact, of which PI3K/AKT pathway keeping sustained activated is one of the most important reasons. This article reviewed the progress of research on the relationship between PI3K/AKT pathway and cisplatin resistance.
Collapse
Affiliation(s)
- Yu Zhang
- Departmen of Cardiothoracic Surgery, Affiliated Hospital of Zunyi Medical College, Zunyi 563099, China
| | - Hongling Lu
- Department of Biochemistry, Zunyi Medical College, Zunyi 563099, China
| | - Gang Xu
- Departmen of Cardiothoracic Surgery, Affiliated Hospital of Zunyi Medical College, Zunyi 563099, China
| |
Collapse
|
22
|
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: 0.7] [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
|
23
|
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: 13.0] [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
|
24
|
Heavey S, O’Byrne KJ, Gately K. Strategies for co-targeting the PI3K/AKT/mTOR pathway in NSCLC. Cancer Treat Rev 2014; 40:445-56. [DOI: 10.1016/j.ctrv.2013.08.006] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 08/11/2013] [Accepted: 08/16/2013] [Indexed: 12/20/2022]
|
25
|
Wong MS, Sidik SM, Mahmud R, Stanslas J. Molecular targets in the discovery and development of novel antimetastatic agents: current progress and future prospects. Clin Exp Pharmacol Physiol 2013; 40:307-19. [PMID: 23534409 DOI: 10.1111/1440-1681.12083] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 03/18/2013] [Accepted: 03/21/2013] [Indexed: 01/08/2023]
Abstract
Tumour invasion and metastasis have been recognized as major causal factors in the morbidity and mortality among cancer patients. Many advances in the knowledge of cancer metastasis have yielded an impressive array of attractive drug targets, including enzymes, receptors and multiple signalling pathways. The present review summarizes the molecular pathogenesis of metastasis and the identification of novel molecular targets used in the discovery of antimetastatic agents. Several promising targets have been highlighted, including receptor tyrosine kinases, effector molecules involved in angiogenesis, matrix metalloproteinases (MMPs), urokinase plasminogen activator, adhesion molecules and their receptors, signalling pathways (e.g. phosphatidylinositol 3-kinase, phospholipase Cγ1, mitogen-activated protein kinases, c-Src kinase, c-Met kinases and heat shock protein. The discovery and development of potential novel therapeutics for each of the targets are also discussed in this review. Among these, the most promising agents that have shown remarkable clinical outcome are anti-angiogenic agents (e.g. bevacizumab). Newer agents, such as c-Met kinase inhibitors, are still undergoing preclinical studies and are yet to have their clinical efficacy proven. Some therapeutics, such as first-generation MMP inhibitors (MMPIs; e.g. marimastat) and more selective versions of them (e.g. prinomastat, tanomastat), have undergone clinical trials. Unfortunately, these drugs produced serious adverse effects that led to the premature termination of their development. In the future, third-generation MMPIs and inhibitors of signalling pathways and adhesion molecules could form valuable novel classes of drugs in the anticancer armamentarium to combat metastasis.
Collapse
Affiliation(s)
- Mei S Wong
- Pharmacotherapeutics Unit, Department of Medicine, University Putra Malaysia, Serdang, Selangor, Malaysia
| | | | | | | |
Collapse
|
26
|
Xie H, Lee MH, Zhu F, Reddy K, Huang Z, Kim DJ, Li Y, Peng C, Lim DY, Kang S, Jung SK, Li X, Li H, Ma W, Lubet RA, Ding J, Bode AM, Dong Z. Discovery of the novel mTOR inhibitor and its antitumor activities in vitro and in vivo. Mol Cancer Ther 2013; 12:950-8. [PMID: 23536724 PMCID: PMC3681861 DOI: 10.1158/1535-7163.mct-12-1241] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The phosphoinositide 3-kinase (PI3-K)/Akt and mTOR signaling pathway plays a critical role in cell survival and proliferation and is often aberrantly activated in many types of cancer. The mTOR kinase protein, one of the key molecules in this pathway, has been shown to be an important target for cancer therapy. In the present study, a ligand docking method was used to screen for novel scaffold mTOR inhibitors. Sixty thousand compounds in the Natural Product Database were screened against the mTOR homologous structure, and 13 commercially available compounds listed in the top-ranked 100 compounds were selected for further examination. Compound [(E)-3-(4-(benzo[d][1,3]dioxol-5-yl)-2-oxobut-3-en-1-yl)- 3-hydroxyindolin-2-one; designated herein as 3HOI-BA-01] was then selected for further study of its antitumor activity. An in vitro study has shown that 3HOI-BA-01 inhibited mTOR kinase activity in a dose-dependent manner by directly binding with mTOR. In a panel of non-small cell lung cancer cells, the compound also attenuated mTOR downstream signaling, including the phosphorylation of p70S6K, S6, and Akt, resulting in G1 cell-cycle arrest and growth inhibition. Results of an in vivo study have shown that intraperitoneal injection of 3HOI-BA-01 in A549 lung tumor-bearing mice effectively suppressed cancer growth without affecting the body weight of the mice. The expression of downstream signaling molecules in the mTOR pathway in tumor tissues was also reduced after 3HOI-BA-01 treatment. Taken together, we identified 3HOI-BA-01 as a novel and effective mTOR inhibitor.
Collapse
Affiliation(s)
- Hua Xie
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Mee-Hyun Lee
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Feng Zhu
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Kanamata Reddy
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Zunnan Huang
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Dong Joon Kim
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Yan Li
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Cong Peng
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Do Young Lim
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Soouk Kang
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Sung Keun Jung
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Xiang Li
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Haitao Li
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Weiya Ma
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Ronald A. Lubet
- National Institutes of Health, National Cancer Institute, MD, USA
| | - Jian Ding
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ann M. Bode
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| |
Collapse
|
27
|
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: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 02/08/2013] [Indexed: 11/06/2022] Open
|
28
|
SASAKI HIDEFUMI, SHITARA MASAYUKI, YOKOTA KEISUKE, HIKOSAKA YU, MORIYAMA SATORU, YANO MOTOKI, FUJII YOSHITAKA. RagD gene expression and NRF2 mutations in lung squamous cell carcinomas. Oncol Lett 2012; 4:1167-1170. [PMID: 23226795 PMCID: PMC3506764 DOI: 10.3892/ol.2012.938] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 06/06/2012] [Indexed: 01/21/2023] Open
Abstract
RagD is a member of the small G protein family, which encodes a recently discovered activator of the mTOR pathway. In vitro, RagD plays an important role in the proliferation of NRF2 gene (NFE2L2) mutated cancer cells. We hypothesized that tumor RagD expression may be correlated with the mutation status of NRF2 in lung cancers. RagD mRNA levels were analyzed by quantitative real-time polymerase chain reaction (qPCR) in 90 surgically-treated lung squamous cell cancer cases, including 14 NRF2 mutation cases, and normalized by β-actin mRNA levels. Mean RagD/β-actin mRNA levels of lung squamous cell carcinoma patients did not differ with age (≤65 vs. >65), Brinkman index (<400 vs. ≥400) or gender. RagD/β-actin mRNA levels were significantly higher in stage III samples (3.204±3.623) compared to stage I samples (1.357±1.560) (P= 0.0039). In addition, higher RagD/β-actin mRNA levels were identified in NRF2 mutant samples (3.107±3.633) compared to wild-type samples (1.774±2.301) (P=0.074). These results suggest that RagD induction by NRF2 activation plays a role in the proliferation of lung squamous cell cancers.
Collapse
Affiliation(s)
- HIDEFUMI SASAKI
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601,
Japan
| | - MASAYUKI SHITARA
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601,
Japan
| | - KEISUKE YOKOTA
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601,
Japan
| | - YU HIKOSAKA
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601,
Japan
| | - SATORU MORIYAMA
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601,
Japan
| | - MOTOKI YANO
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601,
Japan
| | - YOSHITAKA FUJII
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601,
Japan
| |
Collapse
|
29
|
Cao Q, Ju X, Li P, Meng X, Shao P, Cai H, Wang M, Zhang Z, Qin C, Yin C. A functional variant in the MTOR promoter modulates its expression and is associated with renal cell cancer risk. PLoS One 2012; 7:e50302. [PMID: 23209702 PMCID: PMC3508984 DOI: 10.1371/journal.pone.0050302] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 10/18/2012] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The mTOR signaling pathway plays a crucial role in the carcinogenesis of renal cell cancer (RCC). We sought to investigate the influence of genetic variations in the mTOR pathway-related genes on the risk of RCC. METHODS We genotyped 8 potentially functional polymorphisms in AKT1, AKT2, PTEN and MTOR genes using the TaqMan method in a case-control study of 710 RCC patients and 760 cancer-free subjects. Unconditional logistic regression, adjusted for potential confounding factors, was used to assess the risk associations. We then examined the functionality of the important polymorphisms. RESULTS Of the 8 polymorphisms, after adjusting for multiple comparisons, we found a significant association between one variant (rs2295080) in the promoter of MTOR and reduced RCC risk (P = 0.005, OR = 0.74, 95%CI = 0.59-0.91, TG/GG vs. TT). Another variant (rs701848) in the 3'UTR region of PTEN was associated with increased RCC risk (P = 0.014, OR = 1.45, 95%CI = 1.08-1.96, CC vs. TT); however, the association was not significant after adjusting for multiple comparisons. Furthermore, we observed lower MTOR mRNA levels in the presence of the rs2295080G allele in normal renal tissues. The luciferase reporter assay showed that the rs2295080G allele significantly decreased luciferase activity. No other significant association between the selected polymorphisms and RCC risk was observed. CONCLUSIONS Our results suggest that the functional MTOR promoter rs2295080 variant affects RCC susceptibility by modulating the endogenous MTOR expression level. The risk effects and the functional impact of the MTOR rs2295080 variant need further validation.
Collapse
Affiliation(s)
- Qiang Cao
- State Key Laboratory of Reproductive Medicine, Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaobing Ju
- State Key Laboratory of Reproductive Medicine, Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Pu Li
- State Key Laboratory of Reproductive Medicine, Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoxin Meng
- State Key Laboratory of Reproductive Medicine, Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Pengfei Shao
- State Key Laboratory of Reproductive Medicine, Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hongzhou Cai
- State Key Laboratory of Reproductive Medicine, Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Meilin Wang
- Department of Molecular and Genetic Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zhengdong Zhang
- Department of Molecular and Genetic Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chao Qin
- State Key Laboratory of Reproductive Medicine, Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- * E-mail: (CQ); (CY)
| | - Changjun Yin
- State Key Laboratory of Reproductive Medicine, Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- * E-mail: (CQ); (CY)
| |
Collapse
|
30
|
Xie H, Zhu F, Huang Z, Lee MH, Kim DJ, Li X, Lim DY, Jung SK, Kang S, Li H, Reddy K, Wang L, Ma W, Lubet RA, Bode AM, Dong Z. Identification of mammalian target of rapamycin as a direct target of fenretinide both in vitro and in vivo. Carcinogenesis 2012; 33:1814-21. [PMID: 22798378 PMCID: PMC3515856 DOI: 10.1093/carcin/bgs234] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
N-(4-hydroxyphenyl) retinamide (4HPR, fenretinide) is a synthetic
retinoid that has been tested in clinical trials as a cancer therapeutic and
chemopreventive agent. Although 4HPR has been shown to be cytotoxic to many kinds of
cancer cells, the underlying molecular mechanisms are only partially understood. Until
now, no direct cancer-related molecular target has been reported to be involved in the
antitumor activities of 4HPR. Herein, we found that 4HPR inhibited mammalian target of
rapamycin (mTOR) kinase activity by directly binding with mTOR, which suppressed the
activities of both the mTORC1 and the mTORC2 complexes. The predicted binding mode of 4HPR
with mTOR was based on a homology computer model, which showed that 4HPR could bind in the
ATP-binding pocket of the mTOR protein through hydrogen bonds and hydrophobic
interactions. In vitro studies also showed that 4HPR attenuated mTOR
downstream signaling in a panel of non-small-cell lung cancer cells, resulting in growth
inhibition. Moreover, knockdown of mTOR in cancer cells decreased their sensitivity to
4HPR. Results of an in vivo study demonstrated that i.p. injection of
4HPR in A549 lung tumor-bearing mice effectively suppressed cancer growth. The expression
of mTOR downstream signaling molecules in tumor tissues was also decreased after 4HPR
treatment. Taken together, our results are the first to identify mTOR as a direct
antitumor target of 4HPR both in vitro and in vivo,
providing a valuable rationale for guiding the clinical uses of 4HPR.
Collapse
Affiliation(s)
- Hua Xie
- The Hormel Institute, University of Minnesota, Austin, Minnesota 55912-3679, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|