1
|
Deng Q, Huang Y, Zeng J, Li X, Zheng X, Guo L, Shi J, Bai L. Recent advancements in the small-molecule drugs for hepatocellular carcinoma (HCC): Structure-activity relationships, pharmacological activities, and the clinical trials. Biomed Pharmacother 2024; 179:117343. [PMID: 39180795 DOI: 10.1016/j.biopha.2024.117343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND AND AIMS: Hepatocellular carcinoma (HCC) is one of the most common malignancies in the world and the sixth leading cause of cancer death worldwide, and it is urgent to find safe and effective drugs for treatment. As an important therapeutic method, small-molecule drugs are continually being updated to achieve improved therapeutic effects. The purpose of this study was to investigate the structural effects of various FDA-listed small-molecule drugs sorafenib, cabozantinib, lenvatinib, and regorafenib on the corresponding HCC targets and possible structural optimization methods, and to explore the mechanism for identifying potential therapeutic drugs that offer better efficacy and fewer side effects. METHODS The structure-activity relationship, pharmacological actions, and clinical applications of small-molecule drugs were reviewed by referencing MEDLINE, Web of Science, CNKI, and other databases, summarizing and integrating the relevant content. RESULTS The results showed that small-molecule drugs can inhibit HCC primarily by forming hydrogen bonds with Glu885, Asp1046, and Cys919 on the HCC target. HCC can be targeted by inhibiting the activation of multiple pathways, blocking the conduction of downstream signaling, and reducing the formation of tumor blood vessels. In general, small-molecule drugs primarily target four key receptors in HCC: VEGFR, PDGFR, EGFR, and FGFR, to achieve effective treatment. CONCLUSIONS By revealing their structure-activity relationships, pharmacological actions, and clinical trials, small-molecule drugs can offer broad prospects for the development of new medications.
Collapse
Affiliation(s)
- Qichuan Deng
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Jing Zeng
- School of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Xinyu Li
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xianyi Zheng
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Li Guo
- The State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Lan Bai
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; The State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| |
Collapse
|
2
|
Han J, Kim D, Park HJ, Park HJ, Lee SK. Antiproliferative Activity of Gibbosic Acid H through Induction of G 0/G 1 Cell Cycle Arrest and Apoptosis in Human Lung Cancer Cells. J Cancer Prev 2023; 28:201-211. [PMID: 38205360 PMCID: PMC10774477 DOI: 10.15430/jcp.2023.28.4.201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/22/2023] [Accepted: 09/22/2023] [Indexed: 01/12/2024] Open
Abstract
Lung cancer is one of the most common causative cancers worldwide. Particularly, non-small cell lung cancer (NSCLC) accounts for approximately 85% of all lung cancer cases. NSCLC is a serious form of lung cancer that requires prompt diagnosis, and the 5-year survival rate for patients with this disease is only 24%. Gibbosic acid H (GaH), a natural lanostanoid obtained from the Ganoderma species (Ganodermataceae), has antiproliferative activities against colon and lung cancer cells. The aim of the present study was to evaluate the antiproliferative activity of GaH in NSCLC cells and to elucidate the underlying molecular mechanisms. GaH was found to induce G0/G1 cell cycle arrest and autophagy by activating adenosine monophosphate-activated protein kinase in A549 and H1299 cells. The induction of this cell cycle arrest was associated with the downregulation of cyclin E1 and CDK2. Additionally, the induction of autophagy by GaH was correlated with the upregulation of LC3B, beclin-1, and p53 expression. GaH also induced apoptosis by upregulating cleaved caspase-3 and Bax in the lung cancer cells. These findings suggest that GaH has a potential in the growth inhibition of human lung cancer cells.
Collapse
Affiliation(s)
- Jaeho Han
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul, Korea
| | - Donghwa Kim
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul, Korea
| | - Hyen Joo Park
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul, Korea
| | - Hee-Juhn Park
- Department of Pharmaceutical Engineering, Sangji University, Wonju, Korea
| | - Sang Kook Lee
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
3
|
Flieswasser T, Van den Eynde A, Freire Boullosa L, Melis J, Hermans C, Merlin C, Lau HW, Van Audenaerde J, Lardon F, Smits E, Pauwels P, Jacobs J. Targeting CD70 in combination with chemotherapy to enhance the anti-tumor immune effects in non-small cell lung cancer. Oncoimmunology 2023; 12:2192100. [PMID: 36970072 PMCID: PMC10038060 DOI: 10.1080/2162402x.2023.2192100] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
Abstract
Despite the recent emergence of immune checkpoint inhibitors, clinical outcomes of metastatic NSCLC patients remain poor, pointing out the unmet need to develop novel therapies to enhance the anti-tumor immune response in NSCLC. In this regard, aberrant expression of the immune checkpoint molecule CD70 has been reported on many cancer types, including NSCLC. In this study, the cytotoxic and immune stimulatory potential of an antibody-based anti-CD70 (aCD70) therapy was explored as single agent and in combination with docetaxel and cisplatin in NSCLC in vitro and in vivo. Anti-CD70 therapy resulted in NK-mediated killing of NSCLC cells and increased production of pro-inflammatory cytokines by NK cells in vitro. The combination of chemotherapy and anti-CD70 therapy further enhanced NSCLC cell killing. Moreover, in vivo findings showed that the sequential treatment of chemo-immunotherapy resulted in a significant improved survival and delayed tumor growth compared to single agents in Lewis Lung carcinoma-bearing mice. The immunogenic potential of the chemotherapeutic regimen was further highlighted by increased numbers of dendritic cells in the tumor-draining lymph nodes in these tumor-bearing mice after treatment. The sequential combination therapy resulted in enhanced intratumoral infiltration of both T and NK cells, as well as an increase in the ratio of CD8+ T cells over Tregs. The superior effect of the sequential combination therapy on survival was further confirmed in a NCI-H1975-bearing humanized IL15-NSG-CD34+ mouse model. These novel preclinical data demonstrate the potential of combining chemotherapy and aCD70 therapy to enhance anti-tumor immune responses in NSCLC patients.
Collapse
Affiliation(s)
- Tal Flieswasser
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), Wilrijk, Belgium
- Department of Pathology, Antwerp University Hospital, Edegem, Belgium
- CONTACT Tal Flieswasser Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), Wilrijk, Belgium
| | - Astrid Van den Eynde
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), Wilrijk, Belgium
- Department of Pathology, Antwerp University Hospital, Edegem, Belgium
| | - Laurie Freire Boullosa
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), Wilrijk, Belgium
| | - Jöran Melis
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), Wilrijk, Belgium
| | - Christophe Hermans
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), Wilrijk, Belgium
| | - Céline Merlin
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), Wilrijk, Belgium
| | - Ho Wa Lau
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), Wilrijk, Belgium
| | - Jonas Van Audenaerde
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), Wilrijk, Belgium
| | - Filip Lardon
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), Wilrijk, Belgium
| | - Evelien Smits
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), Wilrijk, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Patrick Pauwels
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), Wilrijk, Belgium
- Department of Pathology, Antwerp University Hospital, Edegem, Belgium
| | - Julie Jacobs
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), Wilrijk, Belgium
- Argenx BV, Zwijnaarde, Belgium
| |
Collapse
|
4
|
Padinharayil H, Alappat RR, Joy LM, Anilkumar KV, Wilson CM, George A, Valsala Gopalakrishnan A, Madhyastha H, Ramesh T, Sathiyamoorthi E, Lee J, Ganesan R. Advances in the Lung Cancer Immunotherapy Approaches. Vaccines (Basel) 2022; 10:1963. [PMID: 36423060 PMCID: PMC9693102 DOI: 10.3390/vaccines10111963] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/13/2022] [Accepted: 11/17/2022] [Indexed: 09/19/2023] Open
Abstract
Despite the progress in the comprehension of LC progression, risk, immunologic control, and treatment choices, it is still the primary cause of cancer-related death. LC cells possess a very low and heterogeneous antigenicity, which allows them to passively evade the anticancer defense of the immune system by educating cytotoxic lymphocytes (CTLs), tumor-infiltrating lymphocytes (TILs), regulatory T cells (Treg), immune checkpoint inhibitors (ICIs), and myeloid-derived suppressor cells (MDSCs). Though ICIs are an important candidate in first-line therapy, consolidation therapy, adjuvant therapy, and other combination therapies involving traditional therapies, the need for new predictive immunotherapy biomarkers remains. Furthermore, ICI-induced resistance after an initial response makes it vital to seek and exploit new targets to benefit greatly from immunotherapy. As ICIs, tumor mutation burden (TMB), and microsatellite instability (MSI) are not ideal LC predictive markers, a multi-parameter analysis of the immune system considering tumor, stroma, and beyond can be the future-oriented predictive marker. The optimal patient selection with a proper adjuvant agent in immunotherapy approaches needs to be still revised. Here, we summarize advances in LC immunotherapy approaches with their clinical and preclinical trials considering cancer models and vaccines and the potential of employing immunology to predict immunotherapy effectiveness in cancer patients and address the viewpoints on future directions. We conclude that the field of lung cancer therapeutics can benefit from the use of combination strategies but with comprehension of their limitations and improvements.
Collapse
Affiliation(s)
- Hafiza Padinharayil
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur 680005, Kerala, India
| | - Reema Rose Alappat
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur 680005, Kerala, India
| | - Liji Maria Joy
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur 680005, Kerala, India
| | - Kavya V. Anilkumar
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur 680005, Kerala, India
| | - Cornelia M. Wilson
- Life Sciences Industry Liaison Lab, School of Psychology and Life Sciences, Canterbury Christ Church University, Sandwich CT13 9ND, UK
| | - Alex George
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur 680005, Kerala, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Harishkumar Madhyastha
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | | | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Raja Ganesan
- Institute for Liver and Digestive Diseases, College of Medicine, Hallym University, Chuncheon 24253, Republic of Korea
| |
Collapse
|
5
|
Miladinović M, Vučković L, Klisic A. Comparison of Dako HercepTest and Ventana PATHWAY anti-HER2 (4B5) tests and their correlation with silver in situ hybridization in lung adenocarcinoma. Open Med (Wars) 2021; 16:1503-1512. [PMID: 34708154 PMCID: PMC8500854 DOI: 10.1515/med-2021-0366] [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: 06/05/2021] [Revised: 08/13/2021] [Accepted: 09/14/2021] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Discordant results exist about the role of human epidermal growth factor receptor 2 (HER2) overexpression and/or HER2 amplification in lung adenocarcinoma. We aimed to compare the performance of HercepTest and PATHWAY anti-HER2 (4B5) by correlating immunohistochemistry (IHC) results with silver in situ hybridization (SISH) in adenocarcinoma lung specimens. METHODS A total of 148 surgically resected adenocarcinoma lung specimens were included. RESULTS HER2 overexpression was found in 7.4% patients for HercepTest Dako and in 2.7% patients for 4B5 antibody. The overall coincidence between these two types of antibodies equals 93.9%. The incidence of HER2 amplification in lung adenocarcinoma was 17.6%, of which in 2.7% of the cases high-grade amplification was present. HER2 amplification was present in 90.9% of patients with overexpression of HER2, obtained by using HercepTest Dako and 75% patients using 4B5 antibody. A significant correlation between overexpression of HER2 receptors obtained by HercepTest Dako and 4B5 antibody and HER2 amplification was shown. CONCLUSION The research of the efficiency of targeted molecular therapies with an HER2 antibody may serve as a basis for the introduction of routine HER2 status determination in lung adenocarcinoma, dictating the need for the standardized protocol for HER2 status determination in such pathology.
Collapse
Affiliation(s)
- Mirjana Miladinović
- Department of Pathology, Clinical Center of Montenegro, University of Montenegro-Faculty of Medicine, Podgorica, Montenegro
| | - Ljiljana Vučković
- Department of Pathology, Clinical Center of Montenegro, University of Montenegro-Faculty of Medicine, Podgorica, Montenegro
| | - Aleksandra Klisic
- Center for Laboratory Diagnostics, Primary Health Care Center, University of Montenegro-Faculty of Medicine, Trg Nikole Kovacevica 6, 81000 Podgorica, Montenegro
| |
Collapse
|
6
|
Bai Y, Deng S. A six-long noncoding RNA model predicts prognosis in lung adenocarcinoma. Transl Cancer Res 2020; 9:7505-7518. [PMID: 35117351 PMCID: PMC8799207 DOI: 10.21037/tcr-20-2436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/21/2020] [Indexed: 02/05/2023]
Abstract
Background The incidence and mortality of lung cancer rank first among various malignant tumors. The lack of clear molecular classification and effective individualized treatment greatly limits the treatment benefits of patients. Long non-coding RNAs (lncRNAs) have been demonstrated widely involve in tumor progressing, and been proved easy to detect for occupying majority in transcriptome. However, less work focuses on studying the potency of lncRNAs as molecular typing and prognostic indicator in lung cancer. Methods Based on the 448 lung adenocarcinoma (LUAD) samples and the expression of 14,127 lncRNAs from the Cancer Genome Atlas (TCGA) database, we constructed a co-expression network using weighted gene co-expression network analysis. Then based on the feature module and the overall survival of patients, we constructed a risk score model through Cox proportional hazards regression and verified it with a validation cohort. Finally, according to the median of risk score, the function of this model was enriched. Results We identified a module containing 123 lncRNAs that is related with the prognosis of LUAD. Using univariate and multivariate Cox proportional hazards regression with lasso regression, six lncRNAs were identified to construct a risk score model. The calculation formula shown as follows: risk score = (−0.3057 × EXPVIM-AS1) + (0.9678 × EXPAC092811.1) + (1.0829 × EXPNFIA-AS1) + (−0.3505 × EXPAL035701.1) + (3.9378 × EXPAC079336.4) + (−0.2810 × EXPAL121790.2). Six-lncRNA model can be used as an independent prognostic indicator in LUAD (P<0.001) and the area under the 5-year receiver operating characteristic (ROC) curve is 0.715. Conclusions We developed a six-lncRNA model, which could be used for predicting prognosis and guiding medical treatment in LUAD patients.
Collapse
Affiliation(s)
- Yuquan Bai
- Department of Thoracic Surgery research laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Senyi Deng
- Department of Thoracic Surgery research laboratory, West China Hospital, Sichuan University, Chengdu, China.,Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
A seven-long noncoding RNA signature predicts overall survival for patients with early stage non-small cell lung cancer. Aging (Albany NY) 2019; 10:2356-2366. [PMID: 30205363 PMCID: PMC6188476 DOI: 10.18632/aging.101550] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/06/2018] [Indexed: 12/12/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the most common cancer and cause of cancer-related mortality globally. Increasing evidence suggested that the long non-coding RNAs (lncRNAs) were involved in cancer-related death. To explore the possible prognostic lncRNA biomarkers for NSCLC patients, in the present study, we conducted a comprehensive lncRNA profiling analysis based on 1902 patients from Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) datasets. In the discovery phase, we employed 682 patients from the combination of four GEO datasets (GSE30219, GSE31546, GSE33745 and GSE50081) and conducted a seven-lncRNA formula to predict overall survival (OS). Next, we validated our risk-score formula in two independent datasets, TCGA (n=994) and GSE31210 (n=226). Stratified analysis revealed that the seven-lncRNA signature was significantly associated with OS in stage I patients from both discovery and validation groups (all P<0.001). Additionally, the prognostic value of the seven-lncRNA signature was also found to be favorable in patients carrying wild-type KRAS or EGFR. Bioinformatical analysis suggested that the seven-lncRNA signature affected patients’ prognosis by influencing cell cycle-related pathways. In summary, our findings revealed a seven-lncRNA signature that predicted OS of NSCLC patients, especially in those with early tumor stage and carrying wild-type KRAS or EGFR.
Collapse
|
8
|
Shi W, Feng W, Wang J, Zhai C, Zhang Q, Wang Q, Song Y, Yan X, Chai L, Liu P, Chen Y, Li C, Li M. Clinicopathologic Features and Prognostic Implications of Golgi Phosphoprotein 3 in Non-small Cell Lung Cancer: A Meta-analysis. J Cancer 2019; 10:5754-5763. [PMID: 31737112 PMCID: PMC6843888 DOI: 10.7150/jca.30067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 04/11/2019] [Indexed: 01/11/2023] Open
Abstract
Background: A number of studies have investigated the role of Golgi phosphoprotein-3 (GOLPH3) in the pathogenesis and progression of non-small cell lung cancer (NSCLC). However, the results of previous studies are heterogeneous and controversial. The aim of this meta-analysis was to clarify its association with the clinicopathological characteristics of patients and evaluate the prognostic significance of GOLPH3 in NSCLC. Methods: A systematic search was conducted through PMC, PubMed, Medline, Web of Science, Chinese National Knowledge Infrastructure and Wanfang database. The odds ratio (OR) and hazard ratio (HR) with 95 % CI were calculated by STATA 12.0. Results: 8 qualified studies with a total of 1001 patients with NSCLC were included. Pooled results showed that GOLPH3 was highly expressed in tumor tissues compared with adjacent lung tissues (OR, 7.55), and overexpression of GOLPH3 was significantly correlated with advanced clinical stage (OR, 3.42), poor differentiation of tumor (OR, 1.97) and positive lymph node metastasis (OR, 2.58), but no association with histological type, gender, age or tumor size was found in NSCLC patients. In addition, the pooled HR for overall survival was 1.79 by univariate analysis and 1.91 by multivariate analysis. The pooled HR for progression-free survival was 2.50. Conclusions: GOLPH3 could be a risk factor for progression of NSCLC and might act as a potential prognostic biomarker for NSCLC patients.
Collapse
Affiliation(s)
- Wenhua Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Wei Feng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Cui Zhai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yang Song
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xin Yan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Pengtao Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yuqian Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Cong Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
9
|
Chen H, Wang J, Feng X, Zhu M, Hoffmann S, Hsu A, Qian K, Huang D, Zhao F, Liu W, Zhang H, Cheng Z. Mitochondria-targeting fluorescent molecules for high efficiency cancer growth inhibition and imaging. Chem Sci 2019; 10:7946-7951. [PMID: 31853349 PMCID: PMC6836573 DOI: 10.1039/c9sc01410a] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/25/2019] [Indexed: 01/09/2023] Open
Abstract
Fluorescent mitochondria-accumulating delocalized lipophilic cations (DLCs) for cancer therapy have drawn significant attention in the field of cancer theranostics. One of the most promising fluorescent DLCs, F16, can selectively trigger the apoptosis and necrosis of cancer cells, making it an attractive targeted theranostic drug candidate. However, it suffers from low clinical translation potential, largely due to its inefficient anti-cancer activity (IC50 in the μM range) and poorly understood structure-activity relationship (SAR). In this report, eleven indole-ring substituted F16 derivatives (F16s) were synthesized. Among these derivatives, 5BMF was identified as a highly effective theranostic agent, with in vitro studies showing a low IC50 of ∼50 nM (to H2228 cells) and high cancer to normal cell selectivity index of 225. In vivo studies revealed that tumors treated with 5BMF were significantly suppressed (almost no growth over the treatment period) compared to the PBS treated control group, and also no obvious toxicity to mice was found. In addition, the tumor imaging capability of 5BMF was demonstrated by in vivo fluorescence imaging. Finally, we report for the first time a proposed SAR for F16 DLCs. Our work lays down a solid foundation for translating 5BMF into a novel and highly promising DLC for cancer theranostics.
Collapse
Affiliation(s)
- Hao Chen
- Department of Radiology , The First Hospital of Jilin University , Changchun , 130021 , China .
- Center for Molecular Imaging Research , Shanghai Institute of Materia Medica , Chinese Academy of Sciences , Shanghai , 201203 , China
| | - Jing Wang
- Department of Radiology , The First Hospital of Jilin University , Changchun , 130021 , China .
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Canary Center at Stanford for Cancer Early Detection , Stanford University , California 94305-5344 , USA .
| | - Xin Feng
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Canary Center at Stanford for Cancer Early Detection , Stanford University , California 94305-5344 , USA .
- The College of Veterinary Medicine , Jilin University , Changchun , 130021 , China
| | - Mark Zhu
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Canary Center at Stanford for Cancer Early Detection , Stanford University , California 94305-5344 , USA .
| | - Simon Hoffmann
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Canary Center at Stanford for Cancer Early Detection , Stanford University , California 94305-5344 , USA .
| | - Alex Hsu
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Canary Center at Stanford for Cancer Early Detection , Stanford University , California 94305-5344 , USA .
| | - Kun Qian
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Canary Center at Stanford for Cancer Early Detection , Stanford University , California 94305-5344 , USA .
| | - Daijuan Huang
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Canary Center at Stanford for Cancer Early Detection , Stanford University , California 94305-5344 , USA .
| | - Feng Zhao
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Canary Center at Stanford for Cancer Early Detection , Stanford University , California 94305-5344 , USA .
| | - Wei Liu
- Department of Radiology , The First Hospital of Jilin University , Changchun , 130021 , China .
| | - Huimao Zhang
- Department of Radiology , The First Hospital of Jilin University , Changchun , 130021 , China .
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Canary Center at Stanford for Cancer Early Detection , Stanford University , California 94305-5344 , USA .
| |
Collapse
|
10
|
Li X, Li Z, Wu X, Xiong Z, Yang T, Fu Z, Liu X, Tan X, Zhong F, Wan X, Wang D, Ding X, Yang R, Hou H, Li C, Liu H, Chen K, Jiang H, Zheng M. Deep Learning Enhancing Kinome-Wide Polypharmacology Profiling: Model Construction and Experiment Validation. J Med Chem 2019; 63:8723-8737. [PMID: 31364850 DOI: 10.1021/acs.jmedchem.9b00855] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The kinome-wide virtual profiling of small molecules with high-dimensional structure-activity data is a challenging task in drug discovery. Here, we present a virtual profiling model against a panel of 391 kinases based on large-scale bioactivity data and the multitask deep neural network algorithm. The obtained model yields excellent internal prediction capability with an auROC of 0.90 and consistently outperforms conventional single-task models on external tests, especially for kinases with insufficient activity data. Moreover, more rigorous experimental validations including 1410 kinase-compound pairs showed a high-quality average auROC of 0.75 and confirmed many novel predicted "off-target" activities. Given the verified generalizability, the model was further applied to various scenarios for depicting the kinome-wide selectivity and the association with certain diseases. Overall, the computational model enables us to create a comprehensive kinome interaction network for designing novel chemical modulators or drug repositioning and is of practical value for exploring previously less studied kinases.
Collapse
Affiliation(s)
- Xutong Li
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Zhaojun Li
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,School of Information Management, Dezhou University, 566 West University Road, Dezhou 253023, China
| | - Xiaolong Wu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Zhaoping Xiong
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,School of Life Science and Technology, ShanghaiTech University, 393 Huaxiazhong Road, Shanghai 200031, China
| | - Tianbiao Yang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Zunyun Fu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xiaohong Liu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,School of Life Science and Technology, ShanghaiTech University, 393 Huaxiazhong Road, Shanghai 200031, China
| | - Xiaoqin Tan
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Feisheng Zhong
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xiaozhe Wan
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Dingyan Wang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xiaoyu Ding
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Ruirui Yang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,School of Life Science and Technology, ShanghaiTech University, 393 Huaxiazhong Road, Shanghai 200031, China
| | - Hui Hou
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Chunpu Li
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Hong Liu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Kaixian Chen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,School of Life Science and Technology, ShanghaiTech University, 393 Huaxiazhong Road, Shanghai 200031, China
| | - Hualiang Jiang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,School of Life Science and Technology, ShanghaiTech University, 393 Huaxiazhong Road, Shanghai 200031, China
| | - Mingyue Zheng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| |
Collapse
|
11
|
Wang F, Li Y, Zhang Z, Wang J, Wang J. SHCBP1 regulates apoptosis in lung cancer cells through phosphatase and tensin homolog. Oncol Lett 2019; 18:1888-1894. [PMID: 31423258 PMCID: PMC6614682 DOI: 10.3892/ol.2019.10520] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 05/17/2019] [Indexed: 12/14/2022] Open
Abstract
Src homologous and collagen (SHC) SH2-binding protein 1 (SHCBP1) is a member of the SHC family, and is overexpressed in numerous types of cancer. In addition, apoptosis serves an important role in the development of cancer. The purpose of this study was to examine the effect of SHCBP1 on apoptosis and its potential underlying mechanism in lung cancer cells. Apoptosis was detected by flow cytometry and caspase-3 activity analysis. The expression levels of SHCBP1 and phosphatase and tensin homolog (PTEN) were detected by western blot analysis and reverse transcription-quantitative polymerase chain reaction. Cell viability was determined by MTT assay. The results indicated that SHCBP1 was increased in lung cancer cell lines and lung cancer tissues compared with in normal lung cell lines and tissues. The apoptosis of lung cancer cells was significantly increased by SHCBP1 small interfering RNA (siRNA), as indicated by the increased number of apoptotic cells and enhanced caspase-3 activity. In addition, it was demonstrated that PTEN expression was modulated by SHCBP1 knockdown; silencing of SHCBP1 expression led to a significant increase in PTEN expression. Furthermore, inhibition of PTEN by siRNA reversed the increase in apoptosis induced by SHCBP1 siRNA. These results suggested that SHCBP1 may be upregulated in lung cancer and it may serve a key role in the apoptosis of lung cancer cells; this effect was associated with the expression of PTEN.
Collapse
Affiliation(s)
- Fei Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Yi Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Zhe Zhang
- Department of Neurology, Harbin Children's Hospital, Harbin, Heilongjiang 150086, P.R. China
| | - Jingxin Wang
- Department of Pharmacy, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Jinghao Wang
- Department of Pharmacy, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
12
|
Li K, Zhou F, Zhou Y, Zhang S, Li Q, Li Z, Liu L, Wu G, Meng R. Quinalizarin, a specific CK2 inhibitor, can reduce icotinib resistance in human lung adenocarcinoma cell lines. Int J Mol Med 2019; 44:437-446. [PMID: 31173177 PMCID: PMC6605624 DOI: 10.3892/ijmm.2019.4220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/15/2019] [Indexed: 01/22/2023] Open
Abstract
The abnormal activation of the downstream signaling pathways of epidermal growth factor receptor (EGFR) that are independent of EGFR, contribute to the acquisition of EGFR-tyrosine kinase inhibitor (TKI) resistance in non-small cell lung cancer (NSCLC). The serine/threonine protein kinase casein kinase II (CK2) phosphorylates and modulates several members of the EGFR downstream signaling pathways. Thus, the purpose of the current study was to investigate the effects of the addition of quinalizarin (a specific CK2 inhibitor) to icotinib (an EGFR-TKI) on the proliferation and apoptosis of four NSCLC cell lines and its underlying mechanisms. The human lung adenocarcinoma cell lines HCC827, A549, H1650 and H1975 were employed to represent the EGFR-TKI-sensitive EGFR (EGFR-sensitive) mutation, wild-type EGFR and the EGFR-TKI-resistant EGFR (EGFR-resistant) mutations. The cell viability was determined by the MTT assay. Cell apoptosis was detected by flow cytom-etry using the Annexin V-enhanced green fluorescent protein Apoptosis Detection kit. The level of proteins in the EGFR downstream pathway was observed using a western blot assay. The results showed that the cells with the EGFR-sensitive mutation (HCC827, EGFR E716-A750del) were more sensitive to icotinib compared with those possessing the EGFR wild-type (A549) and the EGFR-resistant mutations (H1650, EGFR E716-A750del and PTEN lost; H1975, EGFR L858R+T790M). Quinalizarin inhibited proliferation and promoted apoptosis in the cells with the EGFR wild-type and resistant mutations, and the addition of quinalizarin to icotinib partially restored their sensitivity to icotinib. Quinalizarin and/or icotinib increased the apoptotic rates in the EGFR-TKI resistant cells, and the combination of these reduced the level of protein downstream of EGFR, including phosphorylated (p-AKT) and p-(ERK). In conclusion, quinalizarin may partially sensitize cells to icotinib by inhibiting proliferation and promoting apoptosis mediated by AKT and ERK in EGFR-TKI resistant NSCLC cell lines.
Collapse
Affiliation(s)
- Ke Li
- Pharmacy Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Fangzheng Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yu Zhou
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Sheng Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Qianwen Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Zhenyu Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Li Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Rui Meng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
13
|
Sippel TR, Johnson AM, Li HY, Hanson D, Nguyen TT, Bullock BL, Poczobutt JM, Kwak JW, Kleczko EK, Weiser-Evans MC, Nemenoff RA. Activation of PPARγ in Myeloid Cells Promotes Progression of Epithelial Lung Tumors through TGFβ1. Mol Cancer Res 2019; 17:1748-1758. [PMID: 31088909 DOI: 10.1158/1541-7786.mcr-19-0236] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/11/2019] [Accepted: 05/10/2019] [Indexed: 12/13/2022]
Abstract
Lung cancer is a heterogeneous disease in which patient-specific treatments are desirable and the development of targeted therapies has been effective. Although mutations in KRAS are frequent in lung adenocarcinoma, there are currently no targeted agents against KRAS. Using a mouse lung adenocarcinoma cell line with a Kras mutation (CMT167), we previously showed that PPARγ activation in lung cancer cells inhibits cell growth in vitro yet promotes tumor progression when activated in myeloid cells of the tumor microenvironment. Here, we report that PPARγ activation in myeloid cells promotes the production of TGFβ1, which, in turn, acts on CMT167 cancer cells to increase migration and induce an epithelial-mesenchymal transition (EMT). Targeting TGFβ1 signaling in CMT167 cells prevented their growth and metastasis in vivo. Similarly, another mouse lung adenocarcinoma cell line with a Kras mutation, LLC, induced TGFβ1 in myeloid cells through PPARγ activation. However, LLC cells are more mesenchymal and did not undergo EMT in response to TGFβ1, nor did LLC require TGFβ1 signaling for metastasis in vivo. Converting CMT167 cells to a mesenchymal phenotype through overexpression of ZEB1 made them unresponsive to TGFβ1 receptor inhibition. The ability of TGFβ1 to induce EMT in lung tumors may represent a critical process in cancer progression. We propose that TGFβ receptor inhibition could provide an additional treatment option for KRAS-mutant epithelial lung tumors.Implications: This study suggests that TGFβ receptor inhibitors may be an effective therapy in a subset of KRAS-mutant patients with non-small cell lung cancer, which show an epithelial phenotype.
Collapse
Affiliation(s)
- Trisha R Sippel
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Amber M Johnson
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Howard Y Li
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado.,Department of Veterans Affairs Medical Center, Denver, Colorado
| | - Dwight Hanson
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Teresa T Nguyen
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Bonnie L Bullock
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Joanna M Poczobutt
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Jeff W Kwak
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Emily K Kleczko
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Mary C Weiser-Evans
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado.,Department of Veterans Affairs Medical Center, Denver, Colorado
| | - Raphael A Nemenoff
- Department of Medicine, University of Colorado, Denver, Aurora, Colorado. .,Department of Veterans Affairs Medical Center, Denver, Colorado
| |
Collapse
|
14
|
Chang WA, Tsai YM, Tsai YC, Wu CY, Chang KF, Lien CT, Hung JY, Hsu YL, Kuo PL. Differential expression profiles of the transcriptome in bone marrow-derived cells in lung cancer revealed by next generation sequencing and bioinformatics. Oncol Lett 2019; 17:4341-4350. [PMID: 30944628 PMCID: PMC6444499 DOI: 10.3892/ol.2019.10085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 02/04/2019] [Indexed: 12/16/2022] Open
Abstract
A pre-metastatic niche (PMN) facilitates cancer metastasis through mobilization and recruitment of bone marrow-derived cells (BMDCs) and associated factors. In bone marrow, hematogenous cells, including osteoclasts, macrophages and lymphocytes, and mesenchymal cells, including mesenchymal stem cells, osteoblasts and adipocytes, are involved in PMN formation. Patients with lung cancer and metastasis have a poor prognosis and shortened median survival time. Bone marrow has been considered fertile ground for dormant and proliferating tumor cells, and mobilizing and recruiting BMDCs and immune cells can establish a PMN. However, the role of BMDCs in PMN formation is not yet fully understood. The present study aimed to investigate the association between BMDCs and PMN in bone marrow tissue samples. The results demonstrated that bone marrow served an important role in lung cancer progression and that eight pathways were potentially involved, including ‘T-cell receptor signaling pathway’, ‘osteoclast differentiation’, ‘MAPK signaling pathway’, ‘VEGF signaling pathway’, ‘leukocyte transendothelial migration’, ‘signaling pathways regulating the pluripotency of stem cells’, ‘oxytocin signaling pathway’ and ‘cell adhesion molecules (CAMs)’. In addition, the present study investigated the role of BMDCs in facilitating lung cancer metastasis. In conclusion, the results from the present study suggested that molecular alterations in gene expression may provide a novel signature in lung cancer, which may aid in the development of novel diagnostic and therapeutic strategies for patients with lung cancer and bone metastasis.
Collapse
Affiliation(s)
- Wei-An Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Ying-Ming Tsai
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C.,School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Yu-Chen Tsai
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Cheng-Ying Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | | | - Chi-Tun Lien
- Department of Internal Medicine, Kaohsiung Municipal United Hospital, Kaohsiung 804, Taiwan, R.O.C
| | - Jen-Yu Hung
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C.,School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Center for Infectious Disease and Cancer Research Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| |
Collapse
|
15
|
Concordance analysis between HER2 immunohistochemistry and in situ hybridization in non-small cell lung cancer. Int J Biol Markers 2018; 33:49-54. [PMID: 28478639 DOI: 10.5301/ijbm.5000271] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE This study aimed to elucidate the concordance between human epidermal growth factor receptor 2 (HER2) immunohistochemistry (IHC) and in situ hybridization (ISH) and the diagnostic accuracy of HER2 IHC in non-small cell lung cancer (NSCLC) through a meta-analysis and diagnostic test accuracy review. METHODS Seven eligible studies and 1,217 patients with NSCLC were included in the review. The concordance between HER2 IHC and ISH was analyzed. To confirm the diagnostic accuracy of HER2 IHC, the sensitivity and specificity were analyzed and the area under the curve (AUC) in the summary receiver operating characteristic (SROC) curve was calculated. RESULTS The concordance rate between HER2 IHC and ISH was 0.795 (95% confidence interval [CI] 0.534-0.929). In the HER2 IHC-negative (score 0/1+) subgroup, the concordance rate was 0.975 (95% CI 0.854-0.996). The concordance rates of the HER2 IHC score 2+ and 3+ subgroups were 0.091 (95% CI 0.039-0.197) and 0.665 (95% CI 0.446-0.830), respectively. In diagnostic test accuracy review, the pooled sensitivity and specificity were 0.67 (95% CI 0.54-0.78) and 0.89 (95% CI 0.87-0.91), respectively. The AUC in the SROC curve was 0.891 and the diagnostic odds ratio was 16.99 (95% CI 5.08-56.76). CONCLUSIONS HER2 IHC was largely in agreement with ISH in cases of HER2 IHC score 0/1+. Because the concordance rates of HER2 IHC score 2/3+ cases were lower than that of HER2 IHC score 0/1+ cases, further studies for detailed analysis criteria for HER2 IHC score 2+ or 3+ are required.
Collapse
|
16
|
Guan ZB, Cao YS, Li Y, Tong WN, Zhuo AS. Knockdown of lncRNA GHET1 suppresses cell proliferation, invasion and LATS1/YAP pathway in non small cell lung cancer. Cancer Biomark 2018; 21:557-563. [PMID: 29286919 DOI: 10.3233/cbm-170431] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND The aim in this study was to explore the role of long non-coding RNA GHET1 in development of non small cell lung cancer (NSCLC). METHODS LncRNA GHET1 expression levels were analyzed by qRT-PCR in tumor tissues and adjacent normal tissues in NSCLC. Measuring the cell proliferation and invasion abilities by CCK8, cell colony formation and transwell invasion assays. Relative protein expression was analyzed by western blot assays. RESULTS Expression of lncRNA GHET1 was notably higher in NSCLC tissues compared with adjacent normal tissues by using qRT-PCR analyses. Higher lncRNA GHET1 expression associated with lymph node metastasis, TNM stage and showed poor outcome in NSCLC patients. Knockdown of lncRNA GHET1 suppressed cell proliferation and invasion capacity and Epithelial-Mesenchymal Transition (EMT) phenomenon of NSCLC cells. Moreover, we demonstrated that knockdown of lncRNA GHET1 suppresses LATS1/YAP pathway signaling pathway by downregulating YAP1 expression in NSCLC cells. CONCLUSIONS GHET1 predicted a poor outcome and acted as a tumor-promoting gene in NSCLC. Thus, inhibition of GHET1 may be a potential target of NSCLC treatment.
Collapse
|
17
|
Zhao D, Zhang T, Hou XM, Ling XL. Knockdown of fascin-1 expression suppresses cell migration and invasion of non-small cell lung cancer by regulating the MAPK pathway. Biochem Biophys Res Commun 2018; 497:694-699. [PMID: 29458026 DOI: 10.1016/j.bbrc.2018.02.134] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 02/15/2018] [Indexed: 01/24/2023]
Abstract
Fascin-1 is a cytoskeletal protein and it can specifically bind to F-actin, it can be abnormally expressed in a variety of solid tumors. Fascin-1 was identified as a factor for poor prognosis in non-small cell lung cancer (NSCLC). However, the relevant molecular mechanisms are not yet fully understood. In this study, the fascin-1 knockdown cells were produced by lentivirus infection, and then cell proliferation, invasion and cell migration assay were used to investigate the role of fascin-1 in NSCLC cells. The MAPK pathway related proteins were determined by western blot. In the current study, lentivirus-mediated fascin-1 knockdown significantly decreased the proliferation of NSCLC cells. Furthermore, fascin-1 silencing partly inhibited cell invasion and migration. Inhibition of fascin-1 decreased the activity of the MAPK pathway. Therefore, targeting fascin-1 may inhibit the growth and metastasis of NSCLC cells, which is a potentially effective therapeutic strategy for treating NSCLC.
Collapse
Affiliation(s)
- Da Zhao
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, People's Republic of China.
| | - Tao Zhang
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, People's Republic of China
| | - Xiao-Ming Hou
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, People's Republic of China
| | - Xiao-Ling Ling
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, People's Republic of China
| |
Collapse
|
18
|
Wang A, Zhao C, Liu X, Su W, Duan G, Xie Z, Chu S, Gao Y. Knockdown of TBRG4 affects tumorigenesis in human H1299 lung cancer cells by regulating DDIT3, CAV1 and RRM2. Oncol Lett 2017; 15:121-128. [PMID: 29387213 PMCID: PMC5769239 DOI: 10.3892/ol.2017.7328] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 08/23/2017] [Indexed: 01/14/2023] Open
Abstract
The transforming growth factor β regulator 4 (TBRG4) gene, located on the 7p14-p13 chromosomal region, is implicated in numerous types of cancer. However, the contribution(s) of TBRG4 in human lung cancer remains unknown. In the present study, the expression of TBRG4 mRNA was investigated in the H1299 lung cancer cell line using the quantitative polymerase chain reaction (qPCR) following the knockdown of TBRG4 by a lentivirus-mediated small interfering RNA (siRNA). Results identified that the expression of TBRG4 within H1299 cells was significantly suppressed (P<0.01) by RNA interference, and 586 genes were differentially expressed following TBRG4 silencing. Ingenuity Pathway Analysis (IPA) revealed that these genes were often associated with infectious diseases, organismal injury, abnormalities and cancer functional networks. Further IPA of these networks revealed that TBRG4 knockdown in H1299 cells deregulated the expression of 21 downstream genes, including the upregulation of DNA damage-inducible transcript 3 (DDIT3), also termed CCAAT/enhancer-binding protein homologous protein, and downregulation of caveolin 1 (CAV1) and ribonucleotide reductase regulatory subunit M2 (RRM2). Results were validated using qPCR and western blotting. Furthermore, immunohistochemical staining of TBRG4 protein identified that expression was markedly increased in carcinoma compared with in normal tissue. In conclusion, TBRG4 serves a role in the tumorigenesis of lung cancer via deregulation of DDIT3, CAV1 and RRM2. The results of the present study may be important in contributing to our understanding of TBRG4 as a target for lung cancer treatment.
Collapse
Affiliation(s)
- Ansheng Wang
- Shandong University School of Medicine, Jinan, Shandong 250100, P.R. China.,Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Chengling Zhao
- Shandong University School of Medicine, Jinan, Shandong 250100, P.R. China
| | - Xuegang Liu
- Department of Cardiac Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Wen Su
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Guixin Duan
- Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Zongyu Xie
- Department of Radiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Shanshan Chu
- Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Yuan Gao
- Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| |
Collapse
|
19
|
Covell DG. A data mining approach for identifying pathway-gene biomarkers for predicting clinical outcome: A case study of erlotinib and sorafenib. PLoS One 2017; 12:e0181991. [PMID: 28792525 PMCID: PMC5549706 DOI: 10.1371/journal.pone.0181991] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 07/10/2017] [Indexed: 12/28/2022] Open
Abstract
A novel data mining procedure is proposed for identifying potential pathway-gene biomarkers from preclinical drug sensitivity data for predicting clinical responses to erlotinib or sorafenib. The analysis applies linear ridge regression modeling to generate a small (N~1000) set of baseline gene expressions that jointly yield quality predictions of preclinical drug sensitivity data and clinical responses. Standard clustering of the pathway-gene combinations from gene set enrichment analysis of this initial gene set, according to their shared appearance in molecular function pathways, yields a reduced (N~300) set of potential pathway-gene biomarkers. A modified method for quantifying pathway fitness is used to determine smaller numbers of over and under expressed genes that correspond with favorable and unfavorable clinical responses. Detailed literature-based evidence is provided in support of the roles of these under and over expressed genes in compound efficacy. RandomForest analysis of potential pathway-gene biomarkers finds average treatment prediction errors of 10% and 22%, respectively, for patients receiving erlotinib or sorafenib that had a favorable clinical response. Higher errors were found for both compounds when predicting an unfavorable clinical response. Collectively these results suggest complementary roles for biomarker genes and biomarker pathways when predicting clinical responses from preclinical data.
Collapse
Affiliation(s)
- David G. Covell
- Information Technology Branch, Developmental Therapeutics Program, National Cancer Institute, Frederick, MD, United States of America
| |
Collapse
|
20
|
Narayanan D, Gani OABSM, Gruber FXE, Engh RA. Data driven polypharmacological drug design for lung cancer: analyses for targeting ALK, MET, and EGFR. J Cheminform 2017; 9:43. [PMID: 29086093 PMCID: PMC5496928 DOI: 10.1186/s13321-017-0229-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 06/18/2017] [Indexed: 12/14/2022] Open
Abstract
Drug design of protein kinase inhibitors is now greatly enabled by thousands of publicly available X-ray structures, extensive ligand binding data, and optimized scaffolds coming off patent. The extensive data begin to enable design against a spectrum of targets (polypharmacology); however, the data also reveal heterogeneities of structure, subtleties of chemical interactions, and apparent inconsistencies between diverse data types. As a result, incorporation of all relevant data requires expert choices to combine computational and informatics methods, along with human insight. Here we consider polypharmacological targeting of protein kinases ALK, MET, and EGFR (and its drug resistant mutant T790M) in non small cell lung cancer as an example. Both EGFR and ALK represent sources of primary oncogenic lesions, while drug resistance arises from MET amplification and EGFR mutation. A drug which inhibits these targets will expand relevant patient populations and forestall drug resistance. Crizotinib co-targets ALK and MET. Analysis of the crystal structures reveals few shared interaction types, highlighting proton-arene and key CH–O hydrogen bonding interactions. These are not typically encoded into molecular mechanics force fields. Cheminformatics analyses of binding data show EGFR to be dissimilar to ALK and MET, but its structure shows how it may be co-targeted with the addition of a covalent trap. This suggests a strategy for the design of a focussed chemical library based on a pan-kinome scaffold. Tests of model compounds show these to be compatible with the goal of ALK, MET, and EGFR polypharmacology.
Collapse
Affiliation(s)
- Dilip Narayanan
- The Norwegian Structural Biology Center, Department of Chemistry, Faculty of Science, UiT The Arctic University of Norway, Tromsø, Norway
| | - Osman A B S M Gani
- The Norwegian Structural Biology Center, Department of Chemistry, Faculty of Science, UiT The Arctic University of Norway, Tromsø, Norway
| | - Franz X E Gruber
- The Norwegian Structural Biology Center, Department of Chemistry, Faculty of Science, UiT The Arctic University of Norway, Tromsø, Norway
| | - Richard A Engh
- The Norwegian Structural Biology Center, Department of Chemistry, Faculty of Science, UiT The Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
21
|
Wang YA, Sun Y, Palmer J, Solomides C, Huang LC, Shyr Y, Dicker AP, Lu B. IGFBP3 Modulates Lung Tumorigenesis and Cell Growth through IGF1 Signaling. Mol Cancer Res 2017; 15:896-904. [PMID: 28330997 DOI: 10.1158/1541-7786.mcr-16-0390] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 11/03/2016] [Accepted: 03/16/2017] [Indexed: 11/16/2022]
Abstract
Insulin-like growth factor binding protein 3 (IGFBP3) modulates cell growth through IGF-dependent and -independent mechanisms. Reports suggest that the serum levels of IGFBP3 are associated with various cancers and that IGFBP3 expression is significantly decreased in cisplatin (CDDP)-resistant lung cancer cells. Based on these findings, we investigated whether Igfbp3 deficiency accelerates mouse lung tumorigenesis and if expression of IGFBP3 enhances CDDP response by focusing on the IGF1 signaling cascade. To this end, an Igfbp3-null mouse model was generated in combination with KrasG12D to compare the tumor burden. Then, IGF-dependent signaling was assessed after expressing wild-type or a mutant IGFBP3 without IGF binding capacity in non-small cell lung cancer (NSCLC) cells. Finally, the treatment response to CDDP chemotherapy was evaluated under conditions of IGFBP3 overexpression. Igfbp3-null mice had increased lung tumor burden (>2-fold) and only half of human lung cancer cells survived after expression of IGFBP3, which corresponded to increased cleaved caspase-3 (10-fold), inactivation of IGF1 and MAPK signaling. In addition, overexpression of IGFBP3 increased susceptibility to CDDP treatment in lung cancer cells. These results, for the first time, demonstrate that IGFBP3 mediates lung cancer progression in a KrasG12D mouse model. Furthermore, overexpression of IGFBP3 induced apoptosis and enhanced cisplatin response in vitro and confirmed that the suppression is in part by blocking IGF1 signaling.Implications: These findings reveal that IGFBP3 is effective in lung cancer cells with high IGF1 signaling activity and imply that relevant biomarkers are essential in selecting lung cancer patients for IGF1-targeted therapy. Mol Cancer Res; 15(7); 896-904. ©2017 AACR.
Collapse
Affiliation(s)
- Yong Antican Wang
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Yunguang Sun
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Joshua Palmer
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | | | - Li-Ching Huang
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yu Shyr
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Adam P Dicker
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Bo Lu
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
22
|
Ransom-Rodríguez A, Blachman-Braun R, Sánchez-García Ramos E, Varela-Prieto J, Rosas-Lezama E, Mercado MÁ. A rare case of choledochal cyst with pancreas divisum: case presentation and literature review. Ann Hepatobiliary Pancreat Surg 2017; 21:52-56. [PMID: 28317046 PMCID: PMC5353911 DOI: 10.14701/ahbps.2017.21.1.52] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 11/13/2016] [Accepted: 11/17/2016] [Indexed: 01/12/2023] Open
Abstract
Choledochal cysts are rare congenital malformations of the bile duct characterized by dilatations of the intrahepatic and/or extrahepatic portion of the biliary tree, they are associated to an anomalous arrangement of the pancreaticobiliary duct. Pancreas divisum results from a fusion failure of the pancreatic buds. The coexistence of pancreas divisum and choledochal cyst in adults has been reported in less than 10 well documented cases. This article presents a case of a 42-year-old Peruvian man with intermittent episodes of abdominal pain, initially diagnosed with choledocholithiasis, who underwent open cholecystectomy. During surgery, a diagnosis of choledochal cyst and pancreas divisum was made, and therefore a hepaticoduodenostomy was performed. The patient was referred to our hospital due to persistence of abdominal pain. After admission, a papillectomy was achieved without further complications. A cyst resection and dismantling of hepaticoduodenostomy with Roux-en-Y was performed 8 years later. During the subsequent 18-month follow-up, the patient remains asymptomatic.
Collapse
Affiliation(s)
- Adrián Ransom-Rodríguez
- Department of Hepatopancreatobiliary Surgery, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.; Facultad Mexicana de Medicina de la Universidad La Salle, Mexico City, Mexico
| | - Ruben Blachman-Braun
- Facultad de Ciencias de la Salud, Universidad Anáhuac México Norte, Edo. de Mexico, Mexico
| | - Emilio Sánchez-García Ramos
- Department of Hepatopancreatobiliary Surgery, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.; Facultad Mexicana de Medicina de la Universidad La Salle, Mexico City, Mexico
| | - Jesús Varela-Prieto
- Department of Hepatopancreatobiliary Surgery, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Erick Rosas-Lezama
- Department of Hepatopancreatobiliary Surgery, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Miguel Ángel Mercado
- Department of Hepatopancreatobiliary Surgery, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
23
|
Wang J, Cheng JX. c-Met inhibition enhances chemosensitivity of human ovarian cancer cells. Clin Exp Pharmacol Physiol 2016; 44:79-87. [PMID: 27658187 DOI: 10.1111/1440-1681.12672] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/16/2016] [Accepted: 09/17/2016] [Indexed: 11/26/2022]
Affiliation(s)
- Jing Wang
- Department of Gynaecology and Obstetrics; The Third Hospital of Hebei Medical University; Shijiazhuang Hebei Province China
| | - Jian-Xin Cheng
- Department of Gynaecology and Obstetrics; The Third Hospital of Hebei Medical University; Shijiazhuang Hebei Province China
| |
Collapse
|
24
|
Silva APS, Coelho PV, Anazetti M, Simioni PU. Targeted therapies for the treatment of non-small-cell lung cancer: Monoclonal antibodies and biological inhibitors. Hum Vaccin Immunother 2016; 13:843-853. [PMID: 27831000 PMCID: PMC5404364 DOI: 10.1080/21645515.2016.1249551] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The usual treatments for patients with non-small-cell lung cancer (NSCLC), such as advanced lung adenocarcinoma, are unspecific and aggressive, and include lung resection, radiotherapy and chemotherapy. Recently, treatment with monoclonal antibodies and biological inhibitors has emerged as an effective alternative, generating effective results with few side effects. In recent years, several clinical trials using monoclonal antibodies presented potential benefits to NSCLC, and 4 of them are already approved for the treatment of NSCLC, such as cetuximab, bevacizumab, nivolumab and pembrolizumab. Also, biological inhibitors are attractive tolls for biological applications. Among the approved inhibitors are crizotinib, erlotinib, afatinib and gefitinib, and side effects are usually mild to intense. Nevertheless, biological molecule treatments are under development, and several new monoclonal antibodies and biological inhibitors are in trial to treat NSCLC. Also under trial study are as follows: anti-epidermal growth factor receptor (EGFR) antibodies (nimotuzumab and ficlatuzumab), anti-IGF 1 receptor (IGF-1R) monoclonal antibody (figitumumab), anti-NR-LU-10 monoclonal antibody (nofetumomab) as well as antibodies directly affecting the cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) molecule (ipilimumab and tremelimumab), to receptor activator of nuclear factor-kappa B ligand (RANKL) (denosumab) or to polymerase enzyme (veliparib and olaparib). Among new inhibitors under investigation are poly-ADP ribose polymerase (PARP) inhibitors (veliparib and olaparib) and phosphatidylinositol 3-kinase (PI3K) inhibitor (buparlisib). However, the success of immunotherapies still requires extensive research and additional controlled trials to evaluate the long-term benefits and side effects.
Collapse
Affiliation(s)
- Ana P S Silva
- a Department of Biomedical Science , Faculty of Americana , Americana , SP , Brazil
| | - Priscila V Coelho
- a Department of Biomedical Science , Faculty of Americana , Americana , SP , Brazil
| | - Maristella Anazetti
- a Department of Biomedical Science , Faculty of Americana , Americana , SP , Brazil.,b Department of Health Science , Faculty DeVry Metrocamp , Campinas , SP , Brazil
| | - Patricia U Simioni
- a Department of Biomedical Science , Faculty of Americana , Americana , SP , Brazil.,c Department of Genetics , Evolution and Bioagents, Institute of Biology, University of Campinas (UNICAMP) , Campinas , SP , Brazil.,d Department of Biochemistry and Microbiology , Institute of Biosciences, Universidade Estadual Paulista, UNESP , Rio Claro , SP , Brazil
| |
Collapse
|
25
|
Dasatinib induces DNA damage and activates DNA repair pathways leading to senescence in non-small cell lung cancer cell lines with kinase-inactivating BRAF mutations. Oncotarget 2016; 7:565-79. [PMID: 26623721 PMCID: PMC4808018 DOI: 10.18632/oncotarget.6376] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 11/14/2015] [Indexed: 12/31/2022] Open
Abstract
Improved therapies are greatly needed for non-small cell lung cancer (NSCLC) that does not harbor targetable kinase mutations or translocations. We previously demonstrated that NSCLC cells that harbor kinase-inactivating BRAF mutations (KIBRAF) undergo senescence when treated with the multitargeted kinase inhibitor dasatinib. Similarly, treatment with dasatinib resulted in a profound and durable response in a patient with KIBRAF NSCLC. However, no canonical pathways explain dasatinib-induced senescence in KIBRAF NSCLC. To investigate the underlying mechanism, we used 2 approaches: gene expression and reverse phase protein arrays. Both approaches showed that DNA repair pathways were differentially modulated between KIBRAF NSCLC cells and those with wild-type (WT) BRAF. Consistent with these findings, dasatinib induced DNA damage and activated DNA repair pathways leading to senescence only in the KIBRAF cells. Moreover, dasatinib-induced senescence was dependent on Chk1 and p21, proteins known to mediate DNA damage-induced senescence. Dasatinib also led to a marked decrease in TAZ but not YAP protein levels. Overexpression of TAZ inhibited dasatinib-induced senescence. To investigate other vulnerabilities in KIBRAF NSCLC cells, we compared the sensitivity of these cells with that of WTBRAF NSCLC cells to 79 drugs and identified a pattern of sensitivity to EGFR and MEK inhibitors in the KIBRAF cells. Clinically approved EGFR and MEK inhibitors, which are better tolerated than dasatinib, could be used to treat KIBRAF NSCLC. Our novel finding that dasatinib induced DNA damage and subsequently activated DNA repair pathways leading to senescence in KIBRAF NSCLC cells represents a unique vulnerability with potential clinical applications.
Collapse
|
26
|
Vanni I, Coco S, Bonfiglio S, Cittaro D, Genova C, Biello F, Mora M, Rossella V, Dal Bello MG, Truini A, Banelli B, Lazarevic D, Alama A, Rijavec E, Barletta G, Grossi F. Whole exome sequencing of independent lung adenocarcinoma, lung squamous cell carcinoma, and malignant peritoneal mesothelioma: A case report. Medicine (Baltimore) 2016; 95:e5447. [PMID: 27902597 PMCID: PMC5134773 DOI: 10.1097/md.0000000000005447] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The presence of multiple primary tumors (MPT) in a single patient has been identified with an increasing frequency. A critical issue is to establish if the second tumor represents an independent primary cancer or a metastasis. Therefore, the assessment of MPT clonal origin might help understand the disease behavior and improve the management/prognosis of the patient.Herein, we report a 73-year-old male smoker who developed 2 primary lung cancers (adenocarcinoma and squamous cell carcinoma) and a malignant peritoneal mesothelioma (PM).Whole exome sequencing (WES) of the 3 tumors and of germline DNA was performed to determine the clonal origin and identify genetic cancer susceptibility.Both lung cancers were characterized by a high mutational rate with distinct mutational profiles and activation of tumor-specific pathways. Conversely, the PM harbored a relative low number of genetic variants and a novel mutation in the WT1 gene that might be involved in the carcinogenesis of nonasbestos-related mesothelioma. Finally, WES of the germinal DNA displayed several single nucleotide polymorphisms in DNA repair genes likely conferring higher cancer susceptibility.Overall, WES did not disclose any somatic genetic variant shared across the 3 tumors, suggesting their clonal independency; however, the carcinogenic effect of smoke combined with a deficiency in DNA repair genes and the patient advanced age might have been responsible for the MPT development. This case highlights the WES importance to define the clonal origin of MPT and susceptibility to cancer.
Collapse
Affiliation(s)
- Irene Vanni
- Lung Cancer Unit, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genova
| | - Simona Coco
- Lung Cancer Unit, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genova
| | - Silvia Bonfiglio
- Centre for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milan
| | - Davide Cittaro
- Centre for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milan
| | - Carlo Genova
- Lung Cancer Unit, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genova
- Department of Internal Medicine and Medical Specialties (DIMI), Università di Genova IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genova
| | - Federica Biello
- Lung Cancer Unit, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genova
| | - Marco Mora
- Department of Pathology, IRCCS AOU San Martino – IST Istituto Nazionale per la Ricerca sul Cancro, Genova
| | - Valeria Rossella
- Centre for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milan
| | - Maria Giovanna Dal Bello
- Lung Cancer Unit, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genova
| | - Anna Truini
- Lung Cancer Unit, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genova
- Department of Internal Medicine and Medical Specialties (DIMI), Università di Genova IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genova
| | - Barbara Banelli
- Laboratory of Tumor Epigenetics, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro and Department of Health Sciences, Università di Genova, Genova, Italy
| | - Dejan Lazarevic
- Centre for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milan
| | - Angela Alama
- Lung Cancer Unit, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genova
| | - Erika Rijavec
- Lung Cancer Unit, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genova
| | - Giulia Barletta
- Lung Cancer Unit, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genova
| | - Francesco Grossi
- Lung Cancer Unit, IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genova
| |
Collapse
|
27
|
Little AC, Sham D, Hristova M, Danyal K, Heppner DE, Bauer RA, Sipsey LM, Habibovic A, van der Vliet A. DUOX1 silencing in lung cancer promotes EMT, cancer stem cell characteristics and invasive properties. Oncogenesis 2016; 5:e261. [PMID: 27694834 PMCID: PMC5117847 DOI: 10.1038/oncsis.2016.61] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 06/30/2016] [Accepted: 08/01/2016] [Indexed: 12/15/2022] Open
Abstract
Dual oxidase 1 (DUOX1) is an oxidant-generating enzyme within the airway epithelium that participates in innate airway host defense and epithelial homeostasis. Recent studies indicate that DUOX1 is suppressed in lung cancers by epigenetic silencing, although the importance of DUOX1 silencing in lung cancer development or progression is unknown. Here we show that loss of DUOX1 expression in a panel of lung cancer cell lines is strongly associated with loss of the epithelial marker E-cadherin. Moreover, RNAi-mediated DUOX1 silencing in lung epithelial cells and the cancer cell line NCI-H292 was found to result in loss of epithelial characteristics/molecular features (altered morphology, reduced barrier function and loss of E-cadherin) and increased mesenchymal features (increased migration, anchorage-independent growth and gain of vimentin/collagen), suggesting a direct contribution of DUOX1 silencing to epithelial-to-mesenchymal transition (EMT), an important feature of metastatic cancer. Conversely, overexpression of DUOX1 in A549 cells was capable of reversing EMT features. DUOX1 silencing in H292 cells also led to enhanced resistance to epidermal growth factor receptor tyrosine kinase inhibitors such as erlotinib, and enhanced levels of cancer stem cell (CSC) markers CD133 and ALDH1. Furthermore, acquired resistance of H292 cells to erlotinib resulted in enhanced EMT and CSC features, as well as loss of DUOX1. Finally, compared with control H292 cells, H292-shDUOX1 cells displayed enhanced invasive features in vitro and in vivo. Collectively, our findings indicate that DUOX1 silencing in lung epithelial cancer cells promotes features of EMT, and may be strongly associated with invasive and metastatic lung cancer.
Collapse
Affiliation(s)
- A C Little
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
- Vermont Lung Center, College of Medicine, University of Vermont, Burlington, VT, USA
| | - D Sham
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
- Vermont Lung Center, College of Medicine, University of Vermont, Burlington, VT, USA
| | - M Hristova
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
- Vermont Lung Center, College of Medicine, University of Vermont, Burlington, VT, USA
| | - K Danyal
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
- Vermont Lung Center, College of Medicine, University of Vermont, Burlington, VT, USA
| | - D E Heppner
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
- Vermont Lung Center, College of Medicine, University of Vermont, Burlington, VT, USA
| | - R A Bauer
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
- Vermont Lung Center, College of Medicine, University of Vermont, Burlington, VT, USA
| | - L M Sipsey
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
- Vermont Lung Center, College of Medicine, University of Vermont, Burlington, VT, USA
| | - A Habibovic
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
- Vermont Lung Center, College of Medicine, University of Vermont, Burlington, VT, USA
| | - A van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
- Vermont Lung Center, College of Medicine, University of Vermont, Burlington, VT, USA
| |
Collapse
|
28
|
Health Resource Utilization in Patients with Advanced Non-Small Cell Lung Cancer Receiving Chemotherapy in China. Clin Drug Investig 2016; 36:77-86. [PMID: 26563152 DOI: 10.1007/s40261-015-0356-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
BACKGROUND AND OBJECTIVES Chemotherapy is the preferred treatment regimen for advanced lung cancer patients. This study investigated the health resources utilized by and medical expenses of patients with non-small cell lung cancer (NSCLC) as well as the influence of various chemotherapy regimens on the final medical costs in China. The aim of this study was to provide physicians with a reference to use as the basis for their choice of treatment. METHODS Data were collected from the Shanghai Chest Hospital's medical charts and billing database. The collected patient information included the baseline characteristics, medical history, chemotherapy regimens, and medical costs, which were used to estimate the health resources utilized by patients and the cost of treatment. RESULTS This study included 328 patients, and the average total medical cost was $US14,165. This cost included drugs, which accounted for as much as 78.91% of the total cost, and chemotherapy drugs, which accounted for 51.58% of total drug expenses. The most frequently utilized chemotherapy drug was carboplatin, and the most expensive chemotherapy drug was erlotinib. In drug combinations, gemcitabine was utilized most frequently, the combination of gemcitabine and paclitaxel was the most expensive, and cisplatin was the least expensive drug. Epidermal growth factor receptor-positive patients were treated with targeted drug therapy (icotinib, erlotinib, and gefitinib). The use of recombinant human endostatin was often combined with a vinorelbine plus cisplatin regimen. Traditional Chinese medicines were the most frequently utilized non-chemotherapy drugs, and these drugs were also the most expensive. CONCLUSIONS The final cost significantly depended on the specific chemotherapy regimen; thus, the rationale and cost of the chemotherapy regimen and adjuvant chemotherapy should be considered in patients with advanced NSCLC.
Collapse
|
29
|
Cytotoxicity of withasteroids: withametelin induces cell cycle arrest at G2/M phase and mitochondria-mediated apoptosis in non-small cell lung cancer A549 cells. Tumour Biol 2016; 37:12579-12587. [PMID: 27382976 DOI: 10.1007/s13277-016-5128-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 06/29/2016] [Indexed: 10/21/2022] Open
Abstract
Considerable interest has been gained by withasteroids because of their structural uniqueness and wide spectrum of biological activities. However, limited systematic studies for proving their cytotoxic potential have so far been reported. Hence, an attempt was made to test the cytotoxicity of six withasteroids viz., withametelin (WM), withaphysalin D, withaphysalin E, 12-deoxywithastramonolide, Withaperuvin B, and physalolactone against A549, HT-29, and MDA-MB-231 cancer cell lines. Significant cytotoxic effect of WM against A549 cells (IC50 value of 6.0 μM), MDA-MB-231 cells (IC50 value of 7.6 μM), and HT-29 cells (IC50 value of 8.2 μM) was observed. Withaperuvin B and physalolactone were found to be effective against MDA-MB-231 cells. The significantly active WM arrested the A549 cells at G2/M phase and downregulated the expression of G2/M regulatory proteins such as cdc2, cyclin B1, and cdc25C. Apoptosis induced by WM in A549 cells was associated with the generation of ROS and depletion of MMP. Furthermore, WM treatment resulted in Bax upregulation, Bcl-2 downregulation, translocation of cytochrome c to mitochondria, activation of caspase-9 and -3, and PARP cleavage corroborating the apoptosis induction through intrinsic apoptotic pathway. Thus, WM possessing broader cytotoxic effect is a promising lead molecule which has the potential to be developed as a new therapeutic agent for NSCLC.
Collapse
|
30
|
Hippo pathway effector YAP inhibition restores the sensitivity of EGFR-TKI in lung adenocarcinoma having primary or acquired EGFR-TKI resistance. Biochem Biophys Res Commun 2016; 474:154-160. [PMID: 27105908 DOI: 10.1016/j.bbrc.2016.04.089] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 04/18/2016] [Indexed: 12/11/2022]
Abstract
The efficacy of EGFR-tyrosine kinase inhibitors (TKIs) is significantly limited by various resistance mechanisms to those drugs. The resistance to EGFR-TKI is largely divided by two classes; acquired resistance after EGFR-TKI treatment, and primary resistance marked by cancer cell's dependence on other oncogene, such as KRAS. YAP has emerged as critical oncogene in conferring drug resistance against targeted therapy. In this study, we evaluated the role of YAP in primary and acquired EGFR-TKI resistance using gefitinib-resistant A549 and PC9 cells and their parental cell lines. Our study revealed that EGFR-TKI resistance is associated with enhanced YAP activity. Notably, YAP activation was independent of the Hippo pathway. We confirmed that AXL is a downstream target of YAP that confers EGFR-TKI resistance. And our results showed that YAP can induce ERK activation in lung adenocarcinoma. The combination of YAP inhibition with EGFR-TKI overcomes primary and acquired EGFR-TKI resistance. We also found increased YAP expression in human lung cancer after acquiring EGFR-TKI resistance. Collectively, we suggest a novel EGFR-TKI resistance mechanism involving YAP activation and suggest targeting YAP and EGFR simultaneously may be a breakthrough treatment of primary and acquired EGFR-TKI resistant lung cancer.
Collapse
|
31
|
Lee JM, Lee SH, Hwang JW, Oh SJ, Kim B, Jung S, Shim SH, Lin PW, Lee SB, Cho MY, Koh YJ, Kim SY, Ahn S, Lee J, Kim KM, Cheong KH, Choi J, Kim KA. Novel strategy for a bispecific antibody: induction of dual target internalization and degradation. Oncogene 2016; 35:4437-46. [PMID: 26853467 DOI: 10.1038/onc.2015.514] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 12/07/2015] [Accepted: 12/14/2015] [Indexed: 02/07/2023]
Abstract
Activation of the extensive cross-talk among the receptor tyrosine kinases (RTKs), particularly ErbB family-Met cross-talk, has emerged as a likely source of drug resistance. Notwithstanding brilliant successes were attained while using small-molecule inhibitors or antibody therapeutics against specific RTKs in multiple cancers over recent decades, a high recurrence rate remains unsolved in patients treated with these targeted inhibitors. It is well aligned with multifaceted properties of cancer and cross-talk and convergence of signaling pathways of RTKs. Thereby many therapeutic interventions have been actively developed to overcome inherent or acquired resistance. To date, no bispecific antibody (BsAb) showed complete depletion of dual RTKs from the plasma membrane and efficient dual degradation. In this manuscript, we report the first findings of a target-specific dual internalization and degradation of membrane RTKs induced by designed BsAbs based on the internalizing monoclonal antibodies and the therapeutic values of these BsAbs. Leveraging the anti-Met mAb able to internalize and degrade by a unique mechanism, we generated the BsAbs for Met/epidermal growth factor receptor (EGFR) and Met/HER2 to induce an efficient EGFR or HER2 internalization and degradation in the presence of Met that is frequently overexpressed in the invasive tumors and involved in the resistance against EGFR- or HER2-targeted therapies. We found that Met/EGFR BsAb ME22S induces dissociation of the Met-EGFR complex from Hsp90, followed by significant degradation of Met and EGFR. By employing patient-derived tumor models we demonstrate therapeutic potential of the BsAb-mediated dual degradation in various cancers.
Collapse
Affiliation(s)
- J M Lee
- Open Innovation Team, Samsung Bioepis Co., Ltd., Incheon, South Korea
| | - S H Lee
- Samsung Biomedical Research Institute, Samsung Advanced Institute of Technology (SAIT), Gyeonggi-do, South Korea
| | - J-W Hwang
- Bioassay Group, Quality Evaluation Team, Samsung Bioepis Co., Ltd., Incheon, South Korea
| | - S J Oh
- Open Innovation Team, Samsung Bioepis Co., Ltd., Incheon, South Korea
| | - B Kim
- Open Innovation Team, Samsung Bioepis Co., Ltd., Incheon, South Korea
| | - S Jung
- Open Innovation Team, Samsung Bioepis Co., Ltd., Incheon, South Korea
| | - S-H Shim
- Samsung Biomedical Research Institute, Samsung Advanced Institute of Technology (SAIT), Gyeonggi-do, South Korea
| | - P W Lin
- Cell Engineering Team, Samsung Bioepis Co., Ltd., Incheon, South Korea
| | - S B Lee
- Cell Engineering Team, Samsung Bioepis Co., Ltd., Incheon, South Korea
| | - M-Y Cho
- Samsung Advanced Institute of Technology (SAIT), Gyeonggi-do, South Korea
| | - Y J Koh
- Samsung Advanced Institute of Technology (SAIT), Gyeonggi-do, South Korea
| | - S Y Kim
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - S Ahn
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - J Lee
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - K-M Kim
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - K H Cheong
- Samsung Advanced Institute of Technology (SAIT), Gyeonggi-do, South Korea
| | - J Choi
- Samsung Biomedical Research Institute, Samsung Advanced Institute of Technology (SAIT), Gyeonggi-do, South Korea
| | - K-A Kim
- Open Innovation Team, Samsung Bioepis Co., Ltd., Incheon, South Korea
| |
Collapse
|
32
|
Poczobutt JM, Nguyen TT, Hanson D, Li H, Sippel TR, Weiser-Evans MCM, Gijon M, Murphy RC, Nemenoff RA. Deletion of 5-Lipoxygenase in the Tumor Microenvironment Promotes Lung Cancer Progression and Metastasis through Regulating T Cell Recruitment. THE JOURNAL OF IMMUNOLOGY 2015; 196:891-901. [PMID: 26663781 DOI: 10.4049/jimmunol.1501648] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 11/09/2015] [Indexed: 12/31/2022]
Abstract
Eicosanoids, including PGs, produced by cyclooxygenases (COX), and leukotrienes, produced by 5-lipoxygenase (5-LO) have been implicated in cancer progression. These molecules are produced by both cancer cells and the tumor microenvironment (TME). We previously reported that both COX and 5-LO metabolites increase during progression in an orthotopic immunocompetent model of lung cancer. Although PGs in the TME have been well studied, less is known regarding 5-LO products produced by the TME. We examined the role of 5-LO in the TME using a model in which Lewis lung carcinoma cells are directly implanted into the lungs of syngeneic WT mice or mice globally deficient in 5-LO (5-LO-KO). Unexpectedly, primary tumor volume and liver metastases were increased in 5-LO-KO mice. This was associated with an ablation of leukotriene (LT) production, consistent with production mainly mediated by the microenvironment. Increased tumor progression was partially reproduced in global LTC4 synthase KO or mice transplanted with LTA4 hydrolase-deficient bone marrow. Tumor-bearing lungs of 5-LO-KO had decreased numbers of CD4 and CD8 T cells compared with WT controls, as well as fewer dendritic cells. This was associated with lower levels of CCL20 and CXL9, which have been implicated in dendritic and T cell recruitment. Depletion of CD8 cells increased tumor growth and eliminated the differences between WT and 5-LO mice. These data reveal an antitumorigenic role for 5-LO products in the microenvironment during lung cancer progression through regulation of T cells and suggest that caution should be used in targeting this pathway in lung cancer.
Collapse
Affiliation(s)
| | - Teresa T Nguyen
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Dwight Hanson
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Howard Li
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045; Veterans Affairs Medical Center, Denver, CO 80220; and
| | - Trisha R Sippel
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Mary C M Weiser-Evans
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045; Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045
| | - Miguel Gijon
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045
| | - Robert C Murphy
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045
| | - Raphael A Nemenoff
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045; Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045
| |
Collapse
|
33
|
Kanthala S, Pallerla S, Jois S. Current and future targeted therapies for non-small-cell lung cancers with aberrant EGF receptors. Future Oncol 2015; 11:865-78. [PMID: 25757687 DOI: 10.2217/fon.14.312] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Expression of the EGF receptors (EGFRs) is abnormally high in many types of cancer, including 25% of lung cancers. Successful treatments target mutations in the EGFR tyrosine kinase domain with EGFR tyrosine kinase inhibitors (TKIs). However, almost all patients develop resistance to this treatment, and acquired resistance to first-generation TKI has prompted the clinical development of a second generation of EGFR TKI. Because of the development of resistance to treatment of TKIs, there is a need to collect genomic information about EGFR levels in non-small-cell lung cancer patients. Herein, we focus on current molecular targets that have therapies available as well as other targets for which therapies will be available in the near future.
Collapse
Affiliation(s)
- Shanthi Kanthala
- Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | | | | |
Collapse
|
34
|
Gandhi S, Chen H, Zhao Y, Dy GK. First-line treatment of advanced ALK-positive non-small-cell lung cancer. LUNG CANCER-TARGETS AND THERAPY 2015; 6:71-82. [PMID: 28210152 PMCID: PMC5217519 DOI: 10.2147/lctt.s63491] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Non-small-cell lung cancer (NSCLC) is one of the leading causes of cancer deaths, both within the US and worldwide. There have been major treatment advances in NSCLC over the past decade with the discovery of molecular drivers of NSCLC, which has ushered in an era of personalized medicine. There are several actionable genetic aberrations in NSCLC, such as epidermal growth factor receptor and anaplastic lymphoma kinase (ALK). In 3%-7% of NSCLC, a chromosomal inversion event in chromosome 2 leads to fusion of a portion of the ALK gene with the echinoderm microtubule-associated protein-like 4 (EML4) gene. The constitutive activation of the ALK fusion oncogene renders it vulnerable to therapeutic intervention. This review focuses on the first-line treatment of advanced ALK-positive NSCLC using ALK inhibitors. Crizotinib was the first agent proven to be efficacious as first-line treatment for ALK-positive NSCLC. However, acquired resistance inevitably develops. The central nervous system is a sanctuary site that represents a common site for disease progression as well. Hence, more potent, selective next-generation ALK inhibitors that are able to cross the blood-brain barrier have been developed for treatment against crizotinib-resistant ALK-positive NSCLC and are also currently being evaluated for first-line therapy as well. In this review, we provide summary of the clinical experience with these drugs in the treatment of ALK-positive NSCLC.
Collapse
Affiliation(s)
- Shipra Gandhi
- Department of Internal Medicine, State University of New York
| | - Hongbin Chen
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Yujie Zhao
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Grace K Dy
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
35
|
Cummings CT, Zhang W, Davies KD, Kirkpatrick GD, Zhang D, DeRyckere D, Wang X, Frye SV, Earp HS, Graham DK. Small Molecule Inhibition of MERTK Is Efficacious in Non-Small Cell Lung Cancer Models Independent of Driver Oncogene Status. Mol Cancer Ther 2015; 14:2014-22. [PMID: 26162689 DOI: 10.1158/1535-7163.mct-15-0116] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 06/25/2015] [Indexed: 12/20/2022]
Abstract
Treatment of non-small cell lung cancer (NSCLC) has been transformed by targeted therapies directed against molecular aberrations specifically activated within an individual patient's tumor. However, such therapies are currently only available against a small number of such aberrations, and new targets and therapeutics are needed. Our laboratory has previously identified the MERTK receptor tyrosine kinase (RTK) as a potential drug target in multiple cancer types, including NSCLC. We have recently developed UNC2025--the first-in-class small molecule inhibitor targeting MERTK with pharmacokinetic properties sufficient for clinical translation. Here, we utilize this compound to further validate the important emerging biologic functions of MERTK in lung cancer pathogenesis, to establish that MERTK can be effectively targeted by a clinically translatable agent, and to demonstrate that inhibition of MERTK is a valid treatment strategy in a wide variety of NSCLC lines independent of their driver oncogene status, including in lines with an EGFR mutation, a KRAS/NRAS mutation, an RTK fusion, or another or unknown driver oncogene. Biochemically, we report the selectivity of UNC2025 for MERTK, and its inhibition of oncogenic downstream signaling. Functionally, we demonstrate that UNC2025 induces apoptosis of MERTK-dependent NSCLC cell lines, while decreasing colony formation in vitro and tumor xenograft growth in vivo in murine models. These findings provide further evidence for the importance of MERTK in NSCLC, and demonstrate that MERTK inhibition by UNC2025 is a feasible, clinically relevant treatment strategy in a wide variety of NSCLC subtypes, which warrants further investigation in clinical trials.
Collapse
Affiliation(s)
- Christopher T Cummings
- Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplantation, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Weihe Zhang
- Center for Integrative Chemical Biology and Drug Discovery and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kurtis D Davies
- Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplantation, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Gregory D Kirkpatrick
- Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplantation, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Dehui Zhang
- Center for Integrative Chemical Biology and Drug Discovery and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Deborah DeRyckere
- Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplantation, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Xiaodong Wang
- Center for Integrative Chemical Biology and Drug Discovery and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Stephen V Frye
- Center for Integrative Chemical Biology and Drug Discovery and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Department of Medicine, UNC Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina
| | - H Shelton Earp
- Department of Medicine, UNC Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina. Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Douglas K Graham
- Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplantation, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
36
|
Yan D, Ge Y, Deng H, Chen W, An G. Gefitinib upregulates death receptor 5 expression to mediate rmhTRAIL-induced apoptosis in Gefitinib-sensitive NSCLC cell line. Onco Targets Ther 2015; 8:1603-10. [PMID: 26170696 PMCID: PMC4498723 DOI: 10.2147/ott.s73731] [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] [Indexed: 01/11/2023] Open
Abstract
Background Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) triggers apoptosis in tumor cells, but when used alone, it is not effective in the treatment of TRAIL-resistant tumors. Some studies have shown that gefitinib interacts with recombinant mutant human TRAIL (rmhTRAIL) to induce high levels of apoptosis in gefitinib-responsive bladder cancer cell lines; however, the molecular mechanisms underlying the anticancer effects are not fully understood. Several reports have shown that the death receptor 5 (DR5) plays an important role in sensitizing cancer cells to apoptosis induced by TRAIL. Therefore, we investigated the effects of the combination of drugs and the expression of the DR5 to analyze the growth of a gefitinib-responsive non-small cell lung cancer cell line PC9, which was treated with rmhTRAIL and gefitinib individually or in combination. Methods Human PC9 non-small cell lung cancer cells harboring an epidermal growth factor receptor mutation were used as a model for the identification of the therapeutic effects of gefitinib alone or in combination with rmhTRAIL, and cytotoxicity was assessed by MTT assays. Cell cycle and apoptosis were investigated using flow cytometry. Moreover, the effects of drugs on DR5, BAX, FLIP, and cleaved-caspase3 proteins expressions were analyzed using Western blot analyses. Finally, quantitative polymerase chain reaction analysis was carried out to assess whether rmhTRAIL and gefitinib modulate the expression of genes related to drug activity. Results Gefitinib and rmhTRAIL synergistically interact to inhibit cell proliferation, and apoptosis assessment demonstrated that associations of drug increased the apoptotic index. rmhTRAIL when used alone downregulated DR5 and upregulated BAX, FLIP, and cleaved-caspase3 proteins expressions. However, results obtained in Western blot analyses demonstrated that the combined treatment-induced cell apoptosis was achieved involving upregulated DR5, cleaved-caspase3, and BAX proteins expression and downregulated FLIP protein expression. Moreover, quantitative polymerase chain reaction showed that gefitinib modulated the expression of targets related to rmhTRAIL activity. Conclusion These results indicate that epidermal growth factor receptor inhibitors enhance rmhTRAIL antitumor activity in the gefitinib-responsive PC9 cell line, and upregulated DR5 expression plays a critical role in activating caspase-signaling apoptotic pathway.
Collapse
Affiliation(s)
- Dong Yan
- Department of Oncology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, People's Republic of China ; Translational Molecular pathology, M.D Anderson Cancer Center, Houston, TX, USA
| | - Yang Ge
- Department of Oncology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Haiteng Deng
- School of Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Wenming Chen
- Department of Hematology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Guangyu An
- Department of Oncology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
37
|
Smith DL, Acquaviva J, Sequeira M, Jimenez JP, Zhang C, Sang J, Bates RC, Proia DA. The HSP90 inhibitor ganetespib potentiates the antitumor activity of EGFR tyrosine kinase inhibition in mutant and wild-type non-small cell lung cancer. Target Oncol 2015; 10:235-45. [PMID: 25077897 PMCID: PMC4457934 DOI: 10.1007/s11523-014-0329-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 07/10/2014] [Indexed: 01/10/2023]
Abstract
Small molecule inhibitors of epidermal growth factor receptor (EGFR) tyrosine kinase activity, such as erlotinib and gefitinib, revolutionized therapy for non-small cell lung cancer (NSCLC) patients whose tumors harbor activating EGFR mutations. However, mechanisms to overcome the invariable development of acquired resistance to such agents, as well as realizing their full clinical potential within the context of wild-type EGFR (WT-EGFR) disease, remain to be established. Here, the antitumor efficacy of targeted EGFR tyrosine kinase inhibitors (TKIs) and the HSP90 inhibitor ganetespib, alone and in combination, were evaluated in NSCLC. Ganetespib potentiated the efficacy of erlotinib in TKI-sensitive, mutant EGFR-driven NCI-HCC827 xenograft tumors, with combination treatment causing significant tumor regressions. In erlotinib-resistant NCI-H1975 xenografts, concurrent administration of ganetespib overcame erlotinib resistance to significantly improve tumor growth inhibition. Ganetespib co-treatment also significantly enhanced antitumor responses to afatinib in the same model. In WT-EGFR cell lines, ganetespib potently reduced cell viability. In NCI-H1666 cells, ganetespib-induced loss of client protein expression, perturbation of oncogenic signaling pathways, and induction of apoptosis translated to robust single-agent activity in vivo. Dual ganetespib/erlotinib therapy induced regressions in NCI-H322 xenograft tumors, indicating that the sensitizing properties of ganetespib for erlotinib were conserved within the WT-EGFR setting. Mechanistically, combined ganetespib/erlotinib exposure stabilized EGFR protein levels in an inactive state and completely abrogated extracellular-signal-regulated kinase (ERK) and AKT signaling activity. Thus, selective HSP90 blockade by ganetespib represents a potentially important complementary strategy to targeted TKI inhibition alone for inducing substantial antitumor responses and overcoming resistance, in both the mutant and WT-EGFR settings.
Collapse
Affiliation(s)
- Donald L. Smith
- Synta Pharmaceuticals Corp, 125 Hartwell Ave, Lexington, MA 02421 USA
| | - Jaime Acquaviva
- Synta Pharmaceuticals Corp, 125 Hartwell Ave, Lexington, MA 02421 USA
| | - Manuel Sequeira
- Synta Pharmaceuticals Corp, 125 Hartwell Ave, Lexington, MA 02421 USA
| | - John-Paul Jimenez
- Synta Pharmaceuticals Corp, 125 Hartwell Ave, Lexington, MA 02421 USA
| | - Chaohua Zhang
- Synta Pharmaceuticals Corp, 125 Hartwell Ave, Lexington, MA 02421 USA
| | - Jim Sang
- Synta Pharmaceuticals Corp, 125 Hartwell Ave, Lexington, MA 02421 USA
| | - Richard C. Bates
- Synta Pharmaceuticals Corp, 125 Hartwell Ave, Lexington, MA 02421 USA
| | - David A. Proia
- Synta Pharmaceuticals Corp, 125 Hartwell Ave, Lexington, MA 02421 USA
| |
Collapse
|
38
|
Xu R, Shang C, Zhao J, Han Y, Liu J, Chen K, Shi W. Activation of M3 muscarinic receptor by acetylcholine promotes non-small cell lung cancer cell proliferation and invasion via EGFR/PI3K/AKT pathway. Tumour Biol 2015; 36:4091-100. [DOI: 10.1007/s13277-014-2911-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 11/27/2014] [Indexed: 11/28/2022] Open
|
39
|
Shen X, Zhuang Z, Zhang Y, Chen Z, Shen L, Pu W, Chen L, Xu Z. JARID1B modulates lung cancer cell proliferation and invasion by regulating p53 expression. Tumour Biol 2015; 36:7133-42. [PMID: 25877751 DOI: 10.1007/s13277-015-3418-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/31/2015] [Indexed: 12/24/2022] Open
Abstract
Although three therapeutic modalities (surgical resection, chemotherapy, and radiotherapy) have been established, long-term survival for lung cancer patients is still generally poor. Until now, the mechanisms of lung cancer genesis remain elusive. The JARID1B is a histone demethylase that has been proposed as oncogene in several types of human cancer, but its clinical significance and functional role in human non-small cell lung cancer (NSCLC) remain unclear. In present study, we found that JARID1B was overexpressed in lung cancer cell lines and lung cancer tissues but not in normal lung tissues. The proliferation and invasive potential of lung cancer cells was significantly increased by ectopic expression of JARID1B. Contrarily, RNA interference targeting JARID1B in lung cancer cells significantly decreased the proliferation and invasive potential of cells. Moreover, we also found that the expression of p53 was modulated by JARID1B. Overexpressed JARID1B cell exhibited greatly decreased p53 expression, whereas silencing of JARID1B expression dramatically increased p53 expression at both the messenger RNA (mRNA) and protein levels. Inhibition of p53 by small interfering RNA (siRNA) reversed the shJARID1B-induced suppression of proliferation and invasion. Our results collectively suggested that JARID1B expressed in lung cancer played a role in lung cancer cells proliferation and invasion, which may be partly associated with the p53 expression.
Collapse
Affiliation(s)
- Xudong Shen
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhixiang Zhuang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yusong Zhang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhigang Chen
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liqin Shen
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wangyang Pu
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Lei Chen
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhonghua Xu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
40
|
Zhao X, Shen L, Feng Y, Yu H, Wu X, Chang J, Shen X, Qiao J, Wang J. Decreased expression of RPS15A suppresses proliferation of lung cancer cells. Tumour Biol 2015; 36:6733-40. [DOI: 10.1007/s13277-015-3371-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 03/24/2015] [Indexed: 12/25/2022] Open
|
41
|
Liang JZ, Li YH, Zhang Y, Wu QN, Wu QL. Expression of ETV6/TEL is associated with prognosis in non-small cell lung cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:2937-45. [PMID: 26045802 PMCID: PMC4440111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 02/23/2015] [Indexed: 06/04/2023]
Abstract
The ETV6/TEL gene is a member of the ETS family of transcription factors that has been mainly studied in hematological diseases. This study provides the first investigation of ETV6 expression in non-small cell lung cancer (NSCLC). In this study, ETV6 expression was immunohistochemically studied in 170 consecutive patients with NSCLC. The association between ETV6 expression and clinicopathological parameters was evaluated. Kaplan-Meier survival analysis and Cox proportional hazards models were used to estimate the effect of ETV6 expression on survival. ETV6 expression was observed in 135 of the 170 (79.4%) patients. ETV6 expression was positive for nuclear staining. From the clinicopathological standpoint, the expression of ETV6 was significantly correlated with age (P=0.014). The overall survival was significantly enhanced in the group with a low expression of ETV6 compared with the group with a high expression of ETV6 (five-year survival rates, 56.53% versus 29.88%; P=0.002), and the same finding was obtained for disease-free survival (five-year survival rates, 52.24% versus 30.47%; P=0.001). Multivariable analysis confirmed that ETV6 expression increased the hazard of death after adjusting for other clinicopathological factors (hazard ratio, 2.002; 95% confidence interval, 1.303-3.074; P=0.002). Our study demonstrated that ETV6 was markedly involved in the development of NSCLC and could serve as a potential prognostic marker for this deadly disease.
Collapse
Affiliation(s)
- Jian-Zhong Liang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer CenterGuangzhou, China
- Department of Pathology, Sun Yat-Sen University Cancer CenterGuangzhou, China
| | - Yuan-Hua Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer CenterGuangzhou, China
- Department of Nuclear Medicine, Sun Yat-Sen University Cancer CenterGuangzhou, China
| | - Yu Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer CenterGuangzhou, China
- Department of Pathology, Sun Yat-Sen University Cancer CenterGuangzhou, China
| | - Qi-Nian Wu
- Department of Pathology, First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou 510120, China
| | - Qiu-Liang Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer CenterGuangzhou, China
- Department of Pathology, Sun Yat-Sen University Cancer CenterGuangzhou, China
| |
Collapse
|
42
|
Wang H, Yan C, Shi X, Zheng J, Deng L, Yang L, Yu F, Yang Y, Shao Y. MicroRNA-575 targets BLID to promote growth and invasion of non-small cell lung cancer cells. FEBS Lett 2015; 589:805-11. [PMID: 25728273 DOI: 10.1016/j.febslet.2015.02.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 01/27/2023]
Abstract
This study was designed to detect miR-575 expression and function in non-small cell lung cancer (NSCLC). A higher expression of miR-575 in NSCLC tissues was observed compared with adjacent non-neoplastic tissues. Furthermore, re-introduction of miR-575 significantly promoted cell proliferation, migration, and invasion in the NSCLC line. Moreover, we showed that BLID is negatively regulated by miR-575 at the posttranscriptional level, via a specific target site within the 3'UTR. Overexpression of BLID counteracted miR-575-induced proliferation and invasion in NSCLC cells. The expression of BLID is frequently downregulated in NSCLC tumors and cell lines and inversely correlates with miR-575 expression. The findings of this study contribute to the current understanding of the functions of miR-575 in NSCLC.
Collapse
Affiliation(s)
- Hongwei Wang
- Department of Respiratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, PR China
| | - Chunhua Yan
- Department of Respiratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, PR China
| | - Xiaodong Shi
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, PR China
| | - Jiaolin Zheng
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, PR China
| | - Lili Deng
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, PR China
| | - Lei Yang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, PR China
| | - Fangfei Yu
- Department of Respiratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, PR China
| | - Yuandi Yang
- Department of Respiratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, PR China
| | - Yuxia Shao
- Department of Respiratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, PR China.
| |
Collapse
|
43
|
Carcereny E, Morán T, Capdevila L, Cros S, Vilà L, de los Llanos Gil M, Remón J, Rosell R. The epidermal growth factor receptor (EGRF) in lung cancer. TRANSLATIONAL RESPIRATORY MEDICINE 2015; 3:1. [PMID: 25810955 PMCID: PMC4366432 DOI: 10.1186/s40247-015-0013-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/16/2015] [Indexed: 12/20/2022]
Abstract
In the last decade, important advances have been made in understanding of cancer biology, particularly non-small-cell lung cancer (NSCLC) with the discovery of oncogenic drivers of the disease. The epidermal growth factor receptor (EGFR) gene and its pathways was the first oncogenic driver discovered to be mutated and treatable in lung cancer. Treatment with EGFR tyrosine kinase inhibitors (TKIs) is the standard of care for molecularly selected EGFR-mutant patients, while its role in unselected lung cancer patients is nowadays controversial. This review will provide an overview of the EGFR pathway and options for its treatment of lung cancer.
Collapse
Affiliation(s)
- Enric Carcereny
- />Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Universitat Autónoma de Barcelona, Department of Medicine, Badalona, Barcelona Spain
| | - Teresa Morán
- />Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Universitat Autónoma de Barcelona, Department of Medicine, Badalona, Barcelona Spain
| | | | - Sara Cros
- />Hospital de Granollers, Barcelona, Spain
| | - Laia Vilà
- />Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Universitat Autónoma de Barcelona, Department of Medicine, Badalona, Barcelona Spain
| | - Maria de los Llanos Gil
- />Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Universitat Autónoma de Barcelona, Department of Medicine, Badalona, Barcelona Spain
| | | | - Rafael Rosell
- />Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Universitat Autónoma de Barcelona, Department of Medicine, Badalona, Barcelona Spain
- />Cancer Biology & Precision Medicine Program, Catalan Institute of Oncology, Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti, Badalona, Barcelona Spain
- />Fundación Molecular Oncology Research, Sabino Arana 5-19, Barcelona, Spain
| |
Collapse
|
44
|
Reis H, Herold T, Ting S, Worm K, Huber U, Christoph DC, Eberhardt WE, Kostbade K, Kasper S, Stamatis G, Welter S, Darwiche K, Karpf-Wissel R, Theegarten D, Schmid KW, Schuler M, Wiesweg M. HER2 expression and markers of phosphoinositide-3-kinase pathway activation define a favorable subgroup of metastatic pulmonary adenocarcinomas. Lung Cancer 2015; 88:34-41. [PMID: 25708529 DOI: 10.1016/j.lungcan.2015.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 01/04/2015] [Accepted: 02/01/2015] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Pulmonary adenocarcinomas (ADC) can be sub-grouped based on dominant oncogenic drivers. EGFR mutations define an entity of metastatic ADC with favorable prognosis and high susceptibility to EGFR tyrosine kinase inhibition. In contrast, the clinical impact of additional ERBB family members in ADC is less defined. To this end we prospectively studied HER2 expression, gene amplification, and mutation in relation to outcome of patients with advanced or metastatic ADC. MATERIALS AND METHODS Diagnostic tumor biopsies from 193 sequential patients with stage III/IV ADC were prospectively studied for HER2 expression by immunohistochemistry (IHC). Cases with IHC scores 2+ or 3+ were analyzed by HER2 chromogenic in situ hybridization (CISH), and sequencing of HER2 exons 20 and 23. Additional prospectively determined biomarkers included PTEN, cMET, pAKT, and pERK expression, KRAS, EGFR, BRAF and PIK3CA mutations, and ALK fluorescence ISH (FISH). RESULTS AND CONCLUSION HER2-IHC was feasible in 176 (91.2%) cases. Of 53 (30%) cases with IHC scores 2+/3+, 45 (85%) could be studied by CISH and 34 (64%) by sequencing. The lower number of HER2-mutational analyses resulted from exhaustion of tumor tissue and DNA following mutational analysis of KRAS, EGFR, BRAF and PIK3CA. HER2 amplification was detected in 4 cases (2.3%), while no mutation was found. HER2 expression correlated with expression of pAKT and cMET. Expression of HER2 and pAKT was associated with favorable overall survival in stage IV disease. HER2-expressing ADC more frequently harbored KRAS mutations, while HER2 expression was absent in all 4 cases with BRAF mutation. HER2-IHC was not predictive of HER2 gene amplification or mutation, which both were rare events in prospectively studied patients with advanced or metastatic ADC. Expression of HER2 and pAKT define a population of patients with stage IV ADC with a distinct disease course, who could benefit from specifically tailored pharmacotherapies.
Collapse
Affiliation(s)
- H Reis
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - T Herold
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany; German Cancer Research Center, 69120 Heidelberg, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - S Ting
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - K Worm
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - U Huber
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - D C Christoph
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - W E Eberhardt
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany; Division of Thoracic Oncology, Ruhrlandklinik, West German Lung Center, University Hospital Essen, 45239 Essen, Germany
| | - K Kostbade
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - S Kasper
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - G Stamatis
- Division of Thoracic Surgery, Ruhrlandklinik, West German Lung Center, University Hospital Essen, 45239 Essen, Germany
| | - S Welter
- Division of Thoracic Surgery, Ruhrlandklinik, West German Lung Center, University Hospital Essen, 45239 Essen, Germany
| | - K Darwiche
- Division of Interventional Pneumology, Ruhrlandklinik, West German Lung Center, University Hospital Essen, 45239 Essen, Germany
| | - R Karpf-Wissel
- Division of Interventional Pneumology, Ruhrlandklinik, West German Lung Center, University Hospital Essen, 45239 Essen, Germany
| | - D Theegarten
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - K W Schmid
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - M Schuler
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany; Division of Thoracic Oncology, Ruhrlandklinik, West German Lung Center, University Hospital Essen, 45239 Essen, Germany.
| | - M Wiesweg
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| |
Collapse
|
45
|
Tian H, Lu JY, Shao C, Huffman KE, Carstens RM, Larsen JE, Girard L, Liu H, Rodriguez-Canales J, Frenkel EP, Wistuba II, Minna JD, Hofmann SL. Systematic siRNA Screen Unmasks NSCLC Growth Dependence by Palmitoyltransferase DHHC5. Mol Cancer Res 2015; 13:784-94. [PMID: 25573953 DOI: 10.1158/1541-7786.mcr-14-0608] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 12/30/2014] [Indexed: 12/11/2022]
Abstract
UNLABELLED Protein S-palmitoylation is a widespread and dynamic posttranslational modification that regulates protein-membrane interactions, protein-protein interactions, and protein stability. A large family of palmitoyl acyl transferases, termed the DHHC family due to the presence of a common catalytic motif, catalyzes S-palmitoylation; the role of these enzymes in cancer is largely unexplored. In this study, an RNAi-based screen targeting all 23 members of the DHHC family was conducted to examine the effects on the growth in non-small cell lung cancer (NSCLC). Interestingly, siRNAs directed against DHHC5 broadly inhibited the growth of multiple NSCLC lines but not normal human bronchial epithelial cell (HBEC) lines. Silencing of DHHC5 by lentivirus-mediated expression of DHHC5 shRNAs dramatically reduced in vitro cell proliferation, colony formation, and cell invasion in a subset of cell lines that were examined in further detail. The phenotypes were restored by transfection of a wild-type DHHC5 plasmid but not by a plasmid expressing a catalytically inactive DHHC5. Tumor xenograft formation was severely inhibited by DHHC5 knockdown and rescued by DHHC5 expression, using both a conventional and tetracycline-inducible shRNA. These data indicate that DHHC5 has oncogenic capacity and contributes to tumor formation in NSCLC, thus representing a potential novel therapeutic target. IMPLICATIONS Inhibitors of DHHC5 enzyme activity may inhibit non-small cell lung cancer growth.
Collapse
Affiliation(s)
- Hui Tian
- The Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jui-Yun Lu
- The Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas. Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chunli Shao
- The Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kenneth E Huffman
- The Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ryan M Carstens
- The Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jill E Larsen
- The Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Luc Girard
- The Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hui Liu
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Jaime Rodriguez-Canales
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Eugene P Frenkel
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - John D Minna
- The Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas. Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas. Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sandra L Hofmann
- The Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas. Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
46
|
Kim Y, Apetri M, Luo B, Settleman JE, Anderson KS. Differential Effects of Tyrosine Kinase Inhibitors on Normal and Oncogenic EGFR Signaling and Downstream Effectors. Mol Cancer Res 2015; 13:765-74. [PMID: 25573954 DOI: 10.1158/1541-7786.mcr-14-0326] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 12/16/2014] [Indexed: 12/11/2022]
Abstract
UNLABELLED Constitutive activation of EGFR due to overexpression or mutation in tumor cells leads to dysregulated downstream cellular signaling pathways. Therefore, EGFR as well as its downstream effectors have been identified as important therapeutic targets. The FDA-approved small-molecule inhibitors of EGFR, gefitinib (Iressa) and erlotinib (Tarceva), are clinically effective in a subset of patients with non-small cell lung cancer (NSCLC) whose tumors harbor activating mutations within the kinase domain of EGFR. The current study examined effects of these drugs in 32D cells expressing native (WT) or oncogenic (L858R) EGFR as well as in cancer cell lines A431 and H3255. Distinct patterns for gefitinib and erlotinib inhibition of EGFR autophosphorylation at individual tyrosines were revealed for wild-type (WT) and L858R EGFR. Phosphorylation of Y845 has been shown to be important in cancer cells and Y1045 phosphorylation is linked to Cbl-mediated ubiquitination and degradation. Dramatic differences were observed by greater potency of these drugs for inhibiting downstream effectors for L858R EGFR including Cbl and STAT5. Selective targeting of Cbl may play a role in oncogene addiction and effects on STAT5 identify features of signaling circuitry for L858R EGFR that contribute to drug sensitivity and clinical efficacy. These data provide new understanding of the EGFR signaling environment and suggest useful paradigms for predicting patient response to EGFR-targeted therapy as well as combination treatments. IMPLICATIONS This study offers fundamental insights for understanding molecular mechanisms of drug sensitivity on oncogenic forms of EGFR and downstream signaling components as well as considerations for further drug optimization and design of combination therapy.
Collapse
Affiliation(s)
- Youngjoo Kim
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut
| | - Mihaela Apetri
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut
| | - BeiBei Luo
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut
| | - Jeffrey E Settleman
- Center for Molecular Therapeutics, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts
| | - Karen S Anderson
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
47
|
Knockdown of PSF1 expression inhibits cell proliferation in lung cancer cells in vitro. Tumour Biol 2014; 36:2163-8. [PMID: 25398693 DOI: 10.1007/s13277-014-2826-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 11/06/2014] [Indexed: 02/06/2023] Open
Abstract
Partner of sld five 1 (PSF1) is a member of the heterotetrameric complex termed GINS. Previous studies have shown that PSF1 is unregulated in several cancer and associated with tumor malignant characters. However, the effects of PSF1 in lung cancer are still unclear. The goal of this study was to investigate the effects of PSF1 on the proliferation capacities of lung cancer. To start with, expression of PSF1 in 22 human lung cancer samples and adjacent non-tumor samples were detected by real-time RT-PCR and Western blotting. Our results showed that PSF1 was overexpressed in lung cancer samples compared to adjacent non-tumor samples. To achieve better insights of PSF1 functions in lung cancer cells, we used PSF1-specific small interfering RNA (siRNA) successfully inhibit the expression of PSF1 in messenger RNA (mRNA) and protein levels. In addition, we used lung cancer cell lines with different p53 gene background (p53 null and p53 wild-type). The results showed that knockdown of PSF1 inhibited cell proliferation and caused cell cycle arrest of lung cancer cells in a p53-independent manner. Our data indicated that PSF1 is functionally involved in lung cancer cell proliferation and is a potential target for lung cancer therapy.
Collapse
|
48
|
Bronte G, Lo Vullo F, Pernice G, Galvano A, Fiorentino E, Cicero G, Bazan V, Rolfo C, Russo A. Farletuzumab for NSCLC: exploiting a well-known metabolic pathway for a new therapeutic strategy. Expert Opin Investig Drugs 2014; 24:125-132. [PMID: 25363562 DOI: 10.1517/13543784.2015.979284] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Introduction: The therapeutic options for NSCLC are limited barring targeted drugs, such as EGFR tyrosine-kinase inhibitors and anaplastic lymphoma kinase inhibitors, for patients bearing oncogenic mutations. Platinum-based chemotherapy remains the best strategy for most patients. New targeted drugs, including mAbs and small molecules, are currently under clinical investigation for treating NSCLC patients. Areas covered: The authors of this article focus on farletuzumab , a mAb targeting folate receptor, which has been studied in ovarian cancer and various other malignancies. In this review, the authors review its potential as therapy for NSCLC, because of the biological rationale provided by the expression of folate receptor α in most of lung adenocarcinoma. The authors provide details of farletuzumab's mechanism of action and discuss the results from completed Phase I and Phase II clinical trials. They also highlight ongoing trials. Expert opinion: There are an increasing number of treatment options for NSCLC and it is hoped that farletuzumab could be added to them. That being said, further evidence for its use with NSCLC patients is still needed. It could have a synergic effect with pemetrexed, because these two drugs have a similar target, namely the folate pathway. This combined action could provide an improved efficacy, although there are some concerns about increased toxicity. However, the authors do note that the combination of farletuzumab with other cytotoxic drugs has not been shown to increase toxicity alone.
Collapse
Affiliation(s)
- Giuseppe Bronte
- University of Palermo, Department of Surgical, Oncological and Oral Sciences , Palermo , Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Hong JY, Park SH, Min HY, Park HJ, Lee SK. Anti-proliferative effects of evodiamine in human lung cancer cells. J Cancer Prev 2014; 19:7-13. [PMID: 25337567 PMCID: PMC4189475 DOI: 10.15430/jcp.2014.19.1.7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 03/18/2014] [Accepted: 03/18/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Evodiamine, a compound isolated from the Evodia rutaecarpa Bentham (Rutaceae), is known to have a potential anti-proliferative activity in human cancer cells. However, the growth inhibitory activity against lung cancer cells and the underlying molecular mechanisms have been poorly determined. The present study was designed to examine the anti-proliferative effect of evodiamine in A549 human lung cancer cells. METHODS A549 cells were treated with the compounds from Evodia rutaecarpa, and the anti-proliferative activity was evaluated by the sulforhodamine B assay. The mechanisms of action for the growth inhibitory activity of evodiamine on A549 human lung cancer cells were evaluated using flow cytometry for cell cycle distribution, and Western blot for assessment of accumulation and phosphorylation of potential target proteins. RESULTS Evodiamine exhibited a potent anti-proliferative activity against A549 human lung cancer cells. Flow cytometric analysis revealed that evodiamine induced cell cycle arrest at G2/M phase and apoptosis in the A549 cells. The cell cycle arrest was well correlated with the inhibition of cyclin B1, cyclin A, cdk2 and p-cdc2 (Tyr15) and increase of p-chk1 (Ser345) and p-chk2 (Thr68). Evodiamine also significantly increased the ratio of Bax/Bcl-2 and decreased procaspase-3, suggesting evodiamine-induced apoptosis via the intrinsic apoptotic pathway. In addition, evodiamine inhibited the expression of p-ERK and ERK. CONCLUSIONS These findings suggest that the anti-proliferative effect of evodiamine was associated in part with the induction of G2/M phase cell cycle arrest and apoptosis, and down-regulation of ERK in human lung cancer cells.
Collapse
Affiliation(s)
- Ji-Young Hong
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - So Hyun Park
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Hye-Young Min
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Hyen Joo Park
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Sang Kook Lee
- College of Pharmacy, Seoul National University, Seoul, Korea
| |
Collapse
|
50
|
Nicoś M, Powrózek T, Krawczyk P, Jarosz B, Pająk B, Sawicki M, Kucharczyk K, Trojanowski T, Milanowski J. Sensitive methods for detection of the S768R substitution in exon 18 of the DDR2 gene in patients with central nervous system metastases of non-small cell lung cancer. Med Oncol 2014; 31:176. [PMID: 25173530 PMCID: PMC4180903 DOI: 10.1007/s12032-014-0176-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 08/09/2014] [Indexed: 11/26/2022]
Abstract
Discoidin death receptor 2 (DDR2) receptor belongs to a DDR family that shows a tyrosine kinase activity. The somatic mutations in DDR2 gene, reported in non-small cell lung cancer (NSCLC), are involved in up-regulation of cells' migration, proliferation and survival. A S768R substitution in DDR2 gene was commonly reported in squamous cell lung carcinoma. Clinical data of patients carrying the DDR2 gene mutation suggest that its presence can be independent of gender and age. The effectiveness of an oral dual-specific (Src and Abl) multikinase inhibitors-dasatinib-was observed in different cell lines and in some NSCLC patients with identified DDR2 mutation. In the present study, we have used three molecular methods (ASP-real-time PCR, ASP-DNA-FLA PCR and direct sequencing) to detect the DDR2 gene mutation in 143 patients with NSCLC metastases to the central nervous system (CNS). The prevalence of the DDR2 gene mutation was correlated with the occurrence of mutations in the EGFR, KRAS, HER2 and BRAF genes. We identified three patients (2.1% of studied group) with DDR2 mutation. The mutation was observed in two patients with low differentiated squamous cell lung cancer and in one patient with adeno-squamous cell carcinoma (ADSCC). In ADSCC patients, DDR2 mutation coexisted with G12C substitution in KRAS gene. According to the current knowledge, examination of the presence of the DDR2 gene mutation in metastatic lesion is the first such report worldwide. The information, that these driver mutations are present in CNS metastases of NSCLC, could broaden therapeutic choices in such group of patients.
Collapse
Affiliation(s)
- Marcin Nicoś
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8, 20-954, Lublin, Poland,
| | | | | | | | | | | | | | | | | |
Collapse
|