1
|
Miao R, Xu Z, Han T, Liu Y, Zhou J, Guo J, Xing Y, Bai Y, He Z, Wu J, Wang W, Hu D. Based on machine learning, CDC20 has been identified as a biomarker for postoperative recurrence and progression in stage I & II lung adenocarcinoma patients. Front Oncol 2024; 14:1351393. [PMID: 39114311 PMCID: PMC11303833 DOI: 10.3389/fonc.2024.1351393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 06/28/2024] [Indexed: 08/10/2024] Open
Abstract
Objective By utilizing machine learning, we can identify genes that are associated with recurrence, invasion, and tumor stemness, thus uncovering new therapeutic targets. Methods To begin, we obtained a gene set related to recurrence and invasion from the GEO database, a comprehensive gene expression database. We then employed the Weighted Gene Co-expression Network Analysis (WGCNA) to identify core gene modules and perform functional enrichment analysis on them. Next, we utilized the random forest and random survival forest algorithms to calculate the genes within the key modules, resulting in the identification of three crucial genes. Subsequently, one of these key genes was selected for prognosis analysis and potential drug screening using the Kaplan-Meier tool. Finally, in order to examine the role of CDC20 in lung adenocarcinoma (LUAD), we conducted a variety of in vitro and in vivo experiments, including wound healing assay, colony formation assays, Transwell migration assays, flow cytometric cell cycle analysis, western blotting, and a mouse tumor model experiment. Results First, we collected a total of 279 samples from two datasets, GSE166722 and GSE31210, to identify 91 differentially expressed genes associated with recurrence, invasion, and stemness in lung adenocarcinoma. Functional enrichment analysis revealed that these key gene clusters were primarily involved in microtubule binding, spindle, chromosomal region, organelle fission, and nuclear division. Next, using machine learning, we identified and validated three hub genes (CDC45, CDC20, TPX2), with CDC20 showing the highest correlation with tumor stemness and limited previous research. Furthermore, we found a close association between CDC20 and clinical pathological features, poor overall survival (OS), progression-free interval (PFI), progression-free survival (PFS), and adverse prognosis in lung adenocarcinoma patients. Lastly, our functional research demonstrated that knocking down CDC20 could inhibit cancer cell migration, invasion, proliferation, cell cycle progression, and tumor growth possibly through the MAPK signaling pathway. Conclusion CDC20 has emerged as a novel biomarker for monitoring treatment response, recurrence, and disease progression in patients with lung adenocarcinoma. Due to its significance, further research studying CDC20 as a potential therapeutic target is warranted. Investigating the role of CDC20 could lead to valuable insights for developing new treatments and improving patient outcomes.
Collapse
Affiliation(s)
- Rui Miao
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, China
- Institute of Precision Medicine (AUST-IPM), Anhui University of Science and Technology, Huainan, China
| | - Zhi Xu
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, China
| | - Tao Han
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, China
| | - Yafeng Liu
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, China
| | - Jiawei Zhou
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, China
| | - Jianqiang Guo
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, China
| | - Yingru Xing
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Department of Clinical Laboratory, Anhui Zhongke Gengjiu Hospital, Hefei, China
| | - Ying Bai
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, China
| | - Zhonglei He
- Institute of Precision Medicine (AUST-IPM), Anhui University of Science and Technology, Huainan, China
- School of Public Health, Anhui University of Science and Technology, Huainan, China
| | - Jing Wu
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, China
| | - Wenxin Wang
- Institute of Precision Medicine (AUST-IPM), Anhui University of Science and Technology, Huainan, China
- School of Public Health, Anhui University of Science and Technology, Huainan, China
| | - Dong Hu
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, China
- Key Laboratory of Industrial Dust Prevention and Control & Occupational Safety and Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, China
| |
Collapse
|
2
|
Zeng H, Ji J, Song X, Huang Y, Li H, Huang J, Ma X. Stemness Related Genes Revealed by Network Analysis Associated With Tumor Immune Microenvironment and the Clinical Outcome in Lung Adenocarcinoma. Front Genet 2020; 11:549213. [PMID: 33193623 PMCID: PMC7525184 DOI: 10.3389/fgene.2020.549213] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 08/24/2020] [Indexed: 02/05/2023] Open
Abstract
Lung adenocarcinoma (LUAD) is one of the leading fatal malignancy with high morbidity and mortality worldwide. However, due to its complicated mechanism and lack of effective clinical therapeutics, early diagnosis and prognosis are still unsatisfactory. Most of the previous studies focused on cancer stem cells (CSCs), the relationship between cancer stemness (stem-like characteristics) and anti-tumor immunity has not been clearly revealed. Therefore, this study aimed to comprehensively analyze the role of cancer stemness and tumor microenvironment (TME) in LUAD using weighted gene co-expression network analysis (WGCNA). We constructed a gene co-expression network, identified key modules, and hub genes, and further explored the relationship between hub gene expression and cancer immunological characteristics through a variety of algorithms, including Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) and Gene Set Enrichment Analysis (GSEA). The hub genes were renamed stemness related genes (SRGs), whose functions were examined at the transcription and protein levels through survival analysis with additional samples, Oncomine database, immunohistochemistry, single cell RNA sequencing (scRNA-seq) and single-sample Gene Set Enrichment Analysis (ssGSEA). Subsequently, Tumor Immune Dysfunction and Exclusion (TIDE) and Connectivity Map (CMap) were implemented for treatment and prognosis analyses. As a result, 15 co-expressed SRGs (CCNA2, CCNB1, CDC20, CDCA5, CDCA8, FEN1, KIF2C, KPNA2, MCM6, NUSAP1, RACGAP1, RRM2, SPAG5, TOP2A, and TPX2) were identified. The overexpression of which was discovered to be associated with reduced immune infiltration in LUAD. It was discovered that there was a general negative correlation between cancer stemness and immunity. The expression of SRGs could probably affect our tumor occurrence, progression, the efficacy of chemotherapy and immunotherapy, and clinical outcomes. In conclusion, the 15 SRGs reported in our study may be used as potential candidate biomarkers for prognostic indicators and therapeutic targets after further validation.
Collapse
Affiliation(s)
- Hao Zeng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu, China.,West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jianrui Ji
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu, China.,West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xindi Song
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yeqian Huang
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Li
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Juan Huang
- Department of Hematology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xuelei Ma
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu, China.,West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Precision Medicine Tumor Boards: Clinical Applicability of Personalized Treatment Concepts in Ovarian Cancer. Cancers (Basel) 2020; 12:cancers12030548. [PMID: 32120793 PMCID: PMC7139570 DOI: 10.3390/cancers12030548] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/14/2020] [Accepted: 02/20/2020] [Indexed: 11/26/2022] Open
Abstract
Background: Treating cancer according to its molecular alterations (i.e., targeted treatment, TT) is the goal of precision medicine tumor boards (PTBs). Their clinical applicability has been evaluated for ovarian cancer patients in this analysis. Methods: All consecutive ovarian cancer patients discussed in a PTB at the Medical University of Vienna, Austria, from April 2015 to April 2019 were included (n = 44). Results: In 38/44 (86%) cases, at least one mutation, deletion or amplification was detected. The most frequently altered genes were p53 (64%), PI3K pathway (18%), KRAS (14%), BRCA1 (11%) and BRCA2 (2%). In 31 patients (70%) a TT was recommended. A total of 12/31 patients (39%) received the recommended therapy. Median time from indication for PTB to TT start was 65 days (15–216). Median time to treatment failure was 2.7 months (0.2–13.2). Clinical benefit rate (CBR) was 42%. Reasons for treatment discontinuation were disease progression (42%), poor performance status (PS > 2; 25%), death (17%) or treatment related side effects (8%). In 61% the TT was not administered—mainly due to PS > 2. Conclusion: Even though a TT recommendation can be derived frequently, clinical applicability remains limited due to poor patients’ general condition after exploitation of standard treatment. However, we observed antitumor activity in a substantial number of heavily pretreated patients.
Collapse
|
4
|
Huang Q, Zhang XW, Ma YS, Lu GX, Xie RT, Yang HQ, Lv ZW, Zhong XM, Liu T, Huang SX, Fu D, Xie C. Up-regulated microRNA-299 corrected with poor prognosis of glioblastoma multiforme patients by targeting ELL2. Jpn J Clin Oncol 2017; 47:590-596. [DOI: 10.1093/jjco/hyw188] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 05/12/2017] [Indexed: 02/02/2023] Open
Affiliation(s)
- Qian Huang
- Department of Burn and Plastic Surgery, People's Hospital of New District Longhua Shenzhen, Shenzhen
| | - Xin-Wen Zhang
- Department of Neurosurgery Surgery, Tongde Hospital of Zhejian Province, Hangzhou
| | - Yu-Shui Ma
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai
| | - Gai-Xia Lu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai
| | - Ru-Ting Xie
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai
| | - Hui-Qiong Yang
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai
| | - Zhong-Wei Lv
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai
| | - Xiao-Ming Zhong
- Department of Radiology, Jiangxi Provincial Tumor Hospital, Nanchang
- Department of Radiology, Ganzhou City People's Hospital, Ganzhou
| | - Tao Liu
- Department of Neurology, People's Hospital of Hainan Province, Haikou
| | - Shi-Xiong Huang
- Department of Neurology, People's Hospital of Hainan Province, Haikou
| | - Da Fu
- Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chun Xie
- Department of Burn and Plastic Surgery, People's Hospital of New District Longhua Shenzhen, Shenzhen
| |
Collapse
|
5
|
Yalak G, Vogel V. Ectokinases as novel cancer markers and drug targets in cancer therapy. Cancer Med 2014; 4:404-14. [PMID: 25504773 PMCID: PMC4380966 DOI: 10.1002/cam4.368] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 09/24/2014] [Accepted: 09/26/2014] [Indexed: 01/13/2023] Open
Abstract
While small-molecule kinase inhibitors became the most prominent anticancer drugs, novel combinatorial strategies need to be developed as the fight against cancer is not yet won. We review emerging literature showing that the release of several ectokinases is significantly upregulated in body fluids from cancer patients and that they leave behind their unique signatures on extracellular matrix (ECM) proteins. Our analysis of proteomic data reveals that fibronectin is heavily phosphorylated in cancer tissues particularly within its growth factor binding sites and on domains that regulate fibrillogenesis. We are thus making the case that cancer is not only a disease of cells but also of the ECM. Targeting extracellular kinases or the extracellular signatures they leave behind might thus create novel opportunities in cancer diagnosis as well as new avenues to interfere with cancer progression and malignancy.
Collapse
Affiliation(s)
- Garif Yalak
- Harvard Medical School/Harvard School of Dental Medicine, Department of Developmental Biology, Harvard University, Boston, Massachusetts, 02115; Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | | |
Collapse
|
6
|
Mansuet-Lupo A, Zouiti F, Alifano M, Tallet A, Charpentier MC, Ducruit V, Devez F, Lemaitre F, Laurent-Puig P, Damotte D, Blons H. Intratumoral distribution of EGFR mutations and copy number in metastatic lung cancer, what impact on the initial molecular diagnosis? J Transl Med 2014; 12:131. [PMID: 24885034 PMCID: PMC4041917 DOI: 10.1186/1479-5876-12-131] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 04/07/2014] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Activating epidermal growth factor receptor (EGFR) mutations characterize a subgroup of non-small-cell lung cancer that benefit from first line EGFR tyrosine kinase inhibitors (EGFR-TKI). However, the existence of polyclonal cell populations may hinder personalized-medicine strategies as patients' screening often depends upon a single tumor-biopsy sample. The purpose of this study is to clarify and to validate in clinical testing conditions the accuracy of EGFR genotyping using different tumor sites and various types of samples (transthoracic, surgical or endoscopic biopsies and cytology specimens). METHODS We conducted a retrospective review of 357 consecutive patients addressed for EGFR mutation screening in accordance with the directive of the European Medicines Agency (stage IV NSCLC). Fifty-seven samples were EGFR mutated and 40 had adequate tumor specimens for analysis on multiple spatially separated sites. Ten wild type samples were also analyzed. A total of 153 and 39 tumor fragments, from mutated and non-mutated cases respectively, were generated to analyze tumor heterogeneity or primary-metastatic discordances. After histological review of all fragments, EGFR genotyping was assessed using the routine diagnostic tools: fragment analysis for insertions and deletions and allele specific TaqMan probes for point mutations. EGFR copy number (CN) was evaluated by qPCR using TaqMan probes. RESULTS The identification of EGFR mutations was independent of localization within primary tumor, of specimen type and consistent between primary and metastases. At the opposite, for half of the samples, tumor loci showed different EGFR copy number that may affect mutation detection cut-off. CONCLUSIONS This is the largest series reporting multiple EGFR testing in Caucasians. It validates the accuracy of EGFR mutation screening from single tumor-biopsy samples before first line EGFR-TKI. The unpredictable variability in EGFR CN and therefore in EGFR wild type/mutant allelic ratio justifies the implementation of sensitive methods to identify patients with EGFR mutated tumors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Hélène Blons
- Université Paris Descartes, Sorbonne, Paris cité, France.
| |
Collapse
|
7
|
Schwaederle M, Parker BA, Schwab RB, Fanta PT, Boles SG, Daniels GA, Bazhenova LA, Subramanian R, Coutinho AC, Ojeda-Fournier H, Datnow B, Webster NJ, Lippman SM, Kurzrock R. Molecular tumor board: the University of California-San Diego Moores Cancer Center experience. Oncologist 2014; 19:631-6. [PMID: 24797821 DOI: 10.1634/theoncologist.2013-0405] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE DNA sequencing tests are enabling physicians to interrogate the molecular profiles of patients' tumors, but most oncologists have not been trained in advanced genomics. We initiated a molecular tumor board to provide expert multidisciplinary input for these patients. MATERIALS AND METHODS A team that included clinicians, basic scientists, geneticists, and bioinformatics/pathway scientists with expertise in various cancer types attended. Molecular tests were performed in a Clinical Laboratory Improvement Amendments environment. RESULTS Patients (n = 34, since December 2012) had received a median of three prior therapies. The median time from physician order to receipt of molecular diagnostic test results was 27 days (range: 14-77 days). Patients had a median of 4 molecular abnormalities (range: 1-14 abnormalities) found by next-generation sequencing (182- or 236-gene panels). Seventy-four genes were involved, with 123 distinct abnormalities. Importantly, no two patients had the same aberrations, and 107 distinct abnormalities were seen only once. Among the 11 evaluable patients whose treatment had been informed by molecular diagnostics, 3 achieved partial responses (progression-free survival of 3.4 months, ≥6.5 months, and 7.6 months). The most common reasons for being unable to act on the molecular diagnostic results were that patients were ineligible for or could not travel to an appropriately targeted clinical trial and/or that insurance would not cover the cognate agents. CONCLUSION Genomic sequencing is revealing complex molecular profiles that differ by patient. Multidisciplinary molecular tumor boards may help optimize management. Barriers to personalized therapy include access to appropriately targeted drugs.
Collapse
Affiliation(s)
- Maria Schwaederle
- Center for Personalized Cancer Therapy, Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, Department of Radiology, School of Medicine, Department of Pathology, School of Medicine, and Division of Endocrinology & Metabolism, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Barbara A Parker
- Center for Personalized Cancer Therapy, Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, Department of Radiology, School of Medicine, Department of Pathology, School of Medicine, and Division of Endocrinology & Metabolism, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Richard B Schwab
- Center for Personalized Cancer Therapy, Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, Department of Radiology, School of Medicine, Department of Pathology, School of Medicine, and Division of Endocrinology & Metabolism, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Paul T Fanta
- Center for Personalized Cancer Therapy, Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, Department of Radiology, School of Medicine, Department of Pathology, School of Medicine, and Division of Endocrinology & Metabolism, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Sarah G Boles
- Center for Personalized Cancer Therapy, Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, Department of Radiology, School of Medicine, Department of Pathology, School of Medicine, and Division of Endocrinology & Metabolism, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Gregory A Daniels
- Center for Personalized Cancer Therapy, Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, Department of Radiology, School of Medicine, Department of Pathology, School of Medicine, and Division of Endocrinology & Metabolism, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Lyudmila A Bazhenova
- Center for Personalized Cancer Therapy, Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, Department of Radiology, School of Medicine, Department of Pathology, School of Medicine, and Division of Endocrinology & Metabolism, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Rupa Subramanian
- Center for Personalized Cancer Therapy, Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, Department of Radiology, School of Medicine, Department of Pathology, School of Medicine, and Division of Endocrinology & Metabolism, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Alice C Coutinho
- Center for Personalized Cancer Therapy, Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, Department of Radiology, School of Medicine, Department of Pathology, School of Medicine, and Division of Endocrinology & Metabolism, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Haydee Ojeda-Fournier
- Center for Personalized Cancer Therapy, Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, Department of Radiology, School of Medicine, Department of Pathology, School of Medicine, and Division of Endocrinology & Metabolism, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Brian Datnow
- Center for Personalized Cancer Therapy, Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, Department of Radiology, School of Medicine, Department of Pathology, School of Medicine, and Division of Endocrinology & Metabolism, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Nicholas J Webster
- Center for Personalized Cancer Therapy, Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, Department of Radiology, School of Medicine, Department of Pathology, School of Medicine, and Division of Endocrinology & Metabolism, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Scott M Lippman
- Center for Personalized Cancer Therapy, Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, Department of Radiology, School of Medicine, Department of Pathology, School of Medicine, and Division of Endocrinology & Metabolism, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, Department of Radiology, School of Medicine, Department of Pathology, School of Medicine, and Division of Endocrinology & Metabolism, Department of Medicine, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
8
|
Cufí S, Bonavia R, Vazquez-Martin A, Oliveras-Ferraros C, Corominas-Faja B, Cuyàs E, Martin-Castillo B, Barrajón-Catalán E, Visa J, Segura-Carretero A, Joven J, Bosch-Barrera J, Micol V, Menendez JA. Silibinin suppresses EMT-driven erlotinib resistance by reversing the high miR-21/low miR-200c signature in vivo. Sci Rep 2014; 3:2459. [PMID: 23963283 PMCID: PMC3748425 DOI: 10.1038/srep02459] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 07/17/2013] [Indexed: 02/07/2023] Open
Abstract
The flavolignan silibinin was studied for its ability to restore drug sensitivity to EGFR-mutant NSCLC xenografts with epithelial-to-mesenchymal transition (EMT)-driven resistance to erlotinib. As a single agent, silibinin significantly decreased the tumor volumes of erlotinib-refractory NSCLC xenografts by approximately 50%. Furthermore, the complete abrogation of tumor growth was observed with the co-treatment of erlotinib and silibinin. Silibinin fully reversed the EMT-related high miR-21/low miR-200c microRNA signature and repressed the mesenchymal markers SNAIL, ZEB, and N-cadherin observed in erlotinib-refractory tumors. Silibinin was sufficient to fully activate a reciprocal mesenchymal-to-epithelial transition (MET) in erlotinib-refractory cells and prevent the highly migratogenic phenotype of erlotinib-resistant NSCLC cells. Given that the various mechanisms of resistance to erlotinib result from EMT, regardless of the EGFR mutation status, a water-soluble, silibinin-rich milk thistle extract might be a suitable candidate therapy for upcoming clinical trials aimed at preventing or reversing NSCLC progression following erlotinib treatment.
Collapse
Affiliation(s)
- Sílvia Cufí
- Metabolism & Cancer Group, Translational Research Laboratory, Catalan Institute of Oncology, Girona, Catalonia, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Corominas-Faja B, Oliveras-Ferraros C, Cuyàs E, Segura-Carretero A, Joven J, Martin-Castillo B, Barrajón-Catalán E, Micol V, Bosch-Barrera J, Menendez JA. Stem cell-like ALDH(bright) cellular states in EGFR-mutant non-small cell lung cancer: a novel mechanism of acquired resistance to erlotinib targetable with the natural polyphenol silibinin. Cell Cycle 2013; 12:3390-404. [PMID: 24047698 DOI: 10.4161/cc.26417] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The enrichment of cancer stem cell (CSC)-like cellular states has not previously been considered to be a causative mechanism in the generalized progression of EGFR-mutant non-small cell lung carcinomas (NSCLC) after an initial response to the EGFR tyrosine kinase inhibitor erlotinib. To explore this possibility, we utilized a pre-clinical model of acquired erlotinib resistance established by growing NSCLC cells containing a TKI-sensitizing EGFR exon 19 deletion (ΔE746-A750) in the continuous presence of high doses of erlotinib. Genome-wide analyses using Agilent 44K Whole Human Genome Arrays were evaluated via bioinformatics analyses through GSEA-based screening of the KEGG pathway database to identify the molecular circuitries that were over-represented in the transcriptomic signatures of erlotinib-refractory cells. The genomic spaces related to erlotinib resistance included a preponderance of cell cycle genes (E2F1, - 2, CDC2, -6) and DNA replication-related genes (MCM4, - 5, - 6, - 7), most of which are associated with early lung development and poor prognosis. In addition, metabolic genes such as ALDH1A3 (a candidate marker for lung cancer cells with CSC-like properties) were identified. Thus, we measured the proportion of erlotinib-resistant cells expressing very high levels of aldehyde dehydrogenase (ALDH) activity attributed to ALDH1/3 isoforms. Using flow cytometry and the ALDEFLUOR® reagent, we confirmed that erlotinib-refractory cell populations contained drastically higher percentages (> 4500%) of ALDH(bright) cells than the parental erlotinib-responsive cells. Notably, strong decreases in the percentages of ALDH(bright) cells were observed following incubation with silibinin, a bioactive flavonolignan that can circumvent erlotinib resistance in vivo. The number of lung cancer spheres was drastically suppressed by silibinin in a dose-dependent manner, thus confirming the ability of this agent to inhibit the self-renewal of erlotinib-refractory CSC-like cells. This report is the first to show that: (1) loss of responsiveness to erlotinib in EGFR-mutant NSCLC can be explained in terms of erlotinib-refractory ALDH(bright) cells, which have been shown to exhibit stem cell-like properties; and (2) erlotinib-refractory ALDH(bright) cells are sensitive to the natural agent silibinin. Our findings highlight the benefit of administration of silibinin in combination with EGFR TKIs to target CSCs and minimize the ability of tumor cells to escape cell death in EGFR-mutant NSCLC patients.
Collapse
Affiliation(s)
- Bruna Corominas-Faja
- Metabolism & Cancer Group; Translational Research Laboratory; Catalan Institute of Oncology; Girona, Catalonia, Spain; Girona Biomedical Research Institute (IDIBGI); Girona, Catalonia, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|