15051
|
SPARC correlates with unfavorable outcome and promotes tumor growth in lung squamous cell carcinoma. Exp Mol Pathol 2019; 110:104276. [PMID: 31233732 DOI: 10.1016/j.yexmp.2019.104276] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 06/17/2019] [Accepted: 06/20/2019] [Indexed: 12/13/2022]
Abstract
Secreted protein acidic and rich in cysteine (SPARC) plays a crucial role in the malignant progression of a number of human cancers. However, the roles of SPARC in lung squamous cell carcinoma (LSCC) remain elusive. In this present study, we first detected SPARC expression and investigated the relationship between SPARC expression and the clinicopathological attributes of LSCC patients. Then we constructed SPARC-overexpression model in LSCC cell line to explore the characteristics of SPARC in LSCC development both in vitro and in vivo. The data demonstrated a remarkably higher level of SPARC in LSCC tissues than in corresponding non-cancerous tissues and elevated SPARC expression was significantly correlated with poor outcome in LSCC patients. Moreover, a serial of phenotypic experiments indicated that SPARC overexpression substantially facilitated the growth and inhibited the apoptosis in LSCC cells and xenografts. Taken together, our results suggest that SPARC is a novel prognostic marker for LSCC prognosis and SPARC significantly promotes LSCC tumorigenesis. Targeting SPARC may provide a novel therapeutic strategy for LSCC management.
Collapse
|
15052
|
Cai Y, Mei J, Xiao Z, Xu B, Jiang X, Zhang Y, Zhu Y. Identification of five hub genes as monitoring biomarkers for breast cancer metastasis in silico. Hereditas 2019; 156:20. [PMID: 31285741 PMCID: PMC6588910 DOI: 10.1186/s41065-019-0096-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 06/12/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Breast cancer is one of the most common endocrine cancers among females worldwide. Distant metastasis of breast cancer is causing an increasing number of breast cancer-related deaths. However, the potential mechanisms of metastasis and candidate biomarkers remain to be further explored. RESULTS The gene expression profiles of GSE102484 were downloaded from the Gene Expression Omnibus (GEO) database. Weighted gene co-expression network analysis (WGCNA) was used to screen for the most potent gene modules associated with the metastatic risk of breast cancer, and a total of 12 modules were identified based on the analysis. In the most significant module (R2 = 0.68), 21 network hub genes (MM > 0.90) were retained for further analyses. Next, protein-protein interaction (PPI) networks were used to further explore the biomarkers with the most interactions in gene modules. According to the PPI networks, five hub genes (TPX2, KIF2C, CDCA8, BUB1B, and CCNA2) were identified as key genes associated with breast cancer progression. Furthermore, the prognostic value and differential expression of these genes were validated based on data from The Cancer Genome Atlas (TCGA) and Kaplan-Meier (KM) Plotter. Receiver operating characteristic (ROC) curve analysis revealed that the mRNA expression levels of these five hub genes showed excellent diagnostic value for breast cancer and adjacent tissues. Moreover, these five hub genes were significantly associated with worse distant metastasis-free survival (DMFS) in the patient cohort based on KM Plotter. CONCLUSION Five hub genes (TPX2, KIF2C, CDCA8, BUB1B, and CCNA2) associated with the risk of distant metastasis were extracted for further research, which might be used as biomarkers to predict distant metastasis of breast cancer.
Collapse
Affiliation(s)
- Yun Cai
- Department of Physiology, Nanjing Medical University, Nanjing, 211166 China
- Department of Bioinformatics, Nanjing Medical University, Nanjing, 211166 China
| | - Jie Mei
- Department of Physiology, Nanjing Medical University, Nanjing, 211166 China
| | - Zhuang Xiao
- Department of Physiology, Nanjing Medical University, Nanjing, 211166 China
| | - Bujie Xu
- Department of Physiology, Nanjing Medical University, Nanjing, 211166 China
| | - Xiaozheng Jiang
- Department of Physiology, Nanjing Medical University, Nanjing, 211166 China
| | - Yongjie Zhang
- Department of Human Anatomy, Nanjing Medical University, Nanjing, 211166 China
- Key Laboratory for Aging & Diseases of Nanjing Medical University, Nanjing Medical University, Nanjing, 211166 China
| | - Yichao Zhu
- Department of Physiology, Nanjing Medical University, Nanjing, 211166 China
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166 China
| |
Collapse
|
15053
|
Zheng Y, Lv P, Wang S, Cai Q, Zhang B, Huo F. LncRNA PLAC2 upregulates p53 to induce hepatocellular carcinoma cell apoptosis. Gene 2019; 712:143944. [PMID: 31233763 DOI: 10.1016/j.gene.2019.143944] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/25/2019] [Accepted: 06/20/2019] [Indexed: 01/07/2023]
Abstract
It is known that lncRNA PLAC2 can inhibit glioma. This study explored the function of PLAC2 in hepatocellular carcinoma (HCC). Our data showed that PLAC2 expression in HCC was not affect by HCV and HBV infection, while PLAC2 levels were significantly lower in HCC tissues comparing to non-cancer tissues. Low PLAC2 levels in HCC tissues were associated with low overall 5-year survival rate. P53 mRNA was also downregulated in HCC and positively correlated with PLAC2. PLAC2 overexpression caused upregulated p53 and increase cancer cell apoptosis. P53 overexpression failed to affect PLAC2. In addition, p53 silencing reduced the effects of PLAC2 overexpression. Therefore, PLAC2 upregulated p53 to mediate cancer cell apoptosis.
Collapse
Affiliation(s)
- Yujian Zheng
- Department of Hepatobiliary Surgery, General Hospital of Southern Theatre Command of PLA, Guangzhou City, Guangdong Province 510010, China.
| | - Pengxiang Lv
- Department of Hepatobiliary Surgery, The 1st People's Hospital of Baiyin, Baiyin, Gansu Province 730900, China
| | - Shaoping Wang
- Department of Hepatobiliary Surgery, General Hospital of Southern Theatre Command of PLA, Guangzhou City, Guangdong Province 510010, China
| | - Qing Cai
- Department of Hepatobiliary Surgery, General Hospital of Southern Theatre Command of PLA, Guangzhou City, Guangdong Province 510010, China
| | - Bao Zhang
- Department of Hepatobiliary Surgery, General Hospital of Southern Theatre Command of PLA, Guangzhou City, Guangdong Province 510010, China
| | - Feng Huo
- Department of Hepatobiliary Surgery, General Hospital of Southern Theatre Command of PLA, Guangzhou City, Guangdong Province 510010, China
| |
Collapse
|
15054
|
Yuan H, Qin Y, Zeng B, Feng Y, Li Y, Xiang T, Ren G. Long noncoding RNA LINC01089 predicts clinical prognosis and inhibits cell proliferation and invasion through the Wnt/β-catenin signaling pathway in breast cancer. Onco Targets Ther 2019; 12:4883-4895. [PMID: 31417284 PMCID: PMC6593691 DOI: 10.2147/ott.s208830] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 05/24/2019] [Indexed: 12/27/2022] Open
Abstract
Background Recently, emerging evidence has indicated crucial roles for long noncoding RNAs (lncRNAs) in breast cancer (BC) development and progression. Our study aimed to investigate the clinical significance of LINC01089 in patients with BC and to determine its biological functions and underlying molecular mechanisms. Materials and methods Correlations between LINC01089 expression and the clinicopathological characteristics of BC patients were assessed using chi-square tests. The Kaplan-Meier method was used to produce survival curves. The clinical risk characteristics associated with the overall survival and recurrence-free survival of patients with BC were estimated using univariate and multivariate Cox regression analyses. Several methods were used to determine the expression profile, biological functions and underlying mechanisms of LINC01089 in BC, including cell proliferation assays, colony formation assays, flow cytometry, transwell assays, wound healing assays, quantitative real-time polymerase chain reaction and Western blotting. Results LINC01089 was downregulated in BC tissues and cell lines. Low LINC01089 expression was significantly correlated with age (P=0.026), lymph node metastasis (P=0.003), and poor prognosis of patients with BC. According to the multivariate Cox regression analysis results, LINC01089 was an independent prognostic indicator of overall survival (P=0.032) and recurrence-free survival (P=0.014). Functional studies revealed significant decreases in the proliferation, migration, and invasion of tumor cells overexpressing LINC01089, and EGF could reverse above effects of LINC01089 on BC cells. Additionally, increased LINC01089 expression promoted apoptosis and cell cycle arrest at G0/G1 phase, accompanied by decreased expression of the key cell cycle regulators CDK4 and CDK6. Loss-of-function assays confirmed partial results. Mechanistically, LINC01089 blocked the Wnt/β-catenin pathway and the expression of downstream target genes by inhibiting β-catenin expression at the transcriptional level. Conclusion Based on our results, LINC01089 functions as a tumor suppressor and potentially represents a novel prognostic indicator and therapeutic target in BC. ![]()
Point your SmartPhone at the code above. If you have a QR code reader the video abstract will appear. Or use: https://youtu.be/v9IJgKoCzJM
Collapse
Affiliation(s)
- Hongfan Yuan
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yi Qin
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Beilei Zeng
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yixiao Feng
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yunhai Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Tingxiu Xiang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
15055
|
Yang F, Jin C, Fu DL, Warshaw AL. Modified FOLFIRINOX for resected pancreatic cancer: Opportunities and challenges. World J Gastroenterol 2019; 25:2839-2845. [PMID: 31249443 PMCID: PMC6589737 DOI: 10.3748/wjg.v25.i23.2839] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/17/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the leading causes of cancer death worldwide. Adjuvant chemotherapy has been developed based on the experiences made with palliative chemotherapy, and advocated to improve long-term survival of patients with this disease. However, the optimal chemotherapeutic regimen remains controversial. Recently, Conroy et al demonstrated the impressive benefits of modified FOLFIRINOX over gemcitabine alone in the multicenter Partenariat de Recherche en Oncologie Digestive 24 (PRODIGE-24) trial. The remarkable results mark a new milestone in treating resectable pancreatic cancer and have now changed the standard of care for this patient population. In this commentary, we discuss an issue of difference of tumor grade between the PRODIGE-24 trial and previous phase III trials. We also discuss potential biomarkers predicting therapeutic response to modified FOLFIRINOX. Finally, we summarize several ongoing clinical trials of replacing part of the FOLFIRINOX regimen with Xeloda/S-1/nanoliposomal irinotecan for pancreatic cancer.
Collapse
Affiliation(s)
- Feng Yang
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Chen Jin
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - De-Liang Fu
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Andrew L Warshaw
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| |
Collapse
|
15056
|
Deng H, Liu W, He T, Hong Z, Yi F, Wei Y, Zhang W. Comparative Efficacy, Safety, and Costs of Sorafenib vs. Sunitinib as First-Line Therapy for Metastatic Renal Cell Carcinoma: A Systematic Review and Meta-Analysis. Front Oncol 2019; 9:479. [PMID: 31293962 PMCID: PMC6598399 DOI: 10.3389/fonc.2019.00479] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/20/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose: Sorafenib and sunitinib are extensively used as first-line medications for metastatic renal cell carcinoma (mRCC). This meta-analysis was conducted to assess the antitumor efficacy, toxicity, and costs of the two drugs among mRCC patients. Materials and methods: PubMed, ScienceDirect, Scopus, Web of Science, Ovid MEDLINE, the Cochrane Library, Embase, and Google Scholar were searched for eligible articles. The endpoints consisted of progression-free survival (PFS), overall survival (OS), objective response rate (ORR), adverse effects (AEs), and per-patient-per-month (PPPM) costs. Results: We included 14 studies with 2,925 patients. Both drugs were valid for treating mRCC with equivalent PFS [hazard ratio (HR) = 0.98, 95% confidence interval (CI): 0.88–1.10, P = 0.74] and disease control rates [DCRs; risk ratio (RR) = 1.03, 95% CI: 0.98–1.08, P = 0.28], but sunitinib had a better OS (HR = 1.10, 95% CI: 1.01–1.20, P = 0.04) and higher ORR (HR = 0.66, 95% CI: 0.45–0.97, P = 0.03) than sorafenib. Furthermore, sunitinib induced more incidences of severe hematologic AEs (anemia, neutropenia, and thrombocytopenia) and stomatitis/mucositis than sorafenib. In the subanalysis, Asian patients treated with sorafenib reported a longer PFS than those treated with sunitinib (HR = 0.87, 95% CI: 0.83–0.90, P = 0.01), and European patients treated with sunitinib had a longer OS than those treated with sorafenib (HR = 1.17, 95% CI: 1.01–1.30, P = 0.04). Moreover, the pooled results of the high-quality studies reported a higher ORR with sunitinib than with sorafenib, and medium-quality studies showed a longer OS with sunitinib than with sorafenib. Conclusions: Sunitinib has more benefits (longer OS and better ORR) than sorafenib as a first-line therapy for mRCC. However, sunitinib has higher toxicity than sorafenib. Sorafenib might be more suitable than sunitinib among Asian patients, and sunitinib might be superior to sorafenib in European patients. Nevertheless, more large-scale, high-quality studies are required.
Collapse
Affiliation(s)
- Huan Deng
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Wenfeng Liu
- Jiangxi Medical College, Nanchang University, Nanchang, China.,Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ting He
- Jiangxi Medical College, Nanchang University, Nanchang, China.,Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhengdong Hong
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fengming Yi
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yiping Wei
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wenxiong Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
15057
|
Lu X, Wang Y, Jiang L, Gao J, Zhu Y, Hu W, Wang J, Ruan X, Xu Z, Meng X, Zhang B, Yan F. A Pre-operative Nomogram for Prediction of Lymph Node Metastasis in Bladder Urothelial Carcinoma. Front Oncol 2019; 9:488. [PMID: 31293963 PMCID: PMC6598397 DOI: 10.3389/fonc.2019.00488] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/23/2019] [Indexed: 12/29/2022] Open
Abstract
The status of lymph node (LN) metastases plays a decisive role in the selection of surgical procedures and post-operative treatment. Several histopathologic features, known as predictors of LN metastasis, are commonly available post-operatively. Medical imaging improved pre-operative diagnosis, but the results are not fully satisfactory due to substantial false positives. Thus, a reliable and robust method for pre-operative assessment of LN status is urgently required. We developed a prediction model in a training set from the TCGA-BLCA cohort including 196 bladder urothelial carcinoma samples with confirmed LN metastasis status. Least absolute shrinkage and selection operator (LASSO) regression was harnessed for dimension reduction, feature selection, and LNM signature building. Multivariable logistic regression was used to develop the prognostic model, incorporating the LNM signature, and a genomic mutation of MLL2, and was presented with a LNM nomogram. The performance of the nomogram was assessed with respect to its calibration, discrimination, and clinical usefulness. Internal validation was evaluated by the testing set from the TCGA cohort and independent validation was assessed by two independent cohorts. The LNM signature, which consisted of 48 selected features, was significantly associated with LN status (p < 0.005 for both the training and testing sets of the TCGA cohort). Predictors contained in the individualized prediction nomogram included the LNM signature and MLL2 mutation status. The model demonstrated good discrimination, with an area under the curve (AUC) of 98.7% (85.3% for testing set) and good calibration with p = 0.973 (0.485 for testing set) in the Hosmer-Lemeshow goodness of fit test. Decision curve analysis demonstrated that the LNM nomogram was clinically useful. This study presents a pre-operative nomogram incorporating a LNM signature and a genomic mutation, which can be conveniently utilized to facilitate pre-operative individualized prediction of LN metastasis in patients with bladder urothelial carcinoma.
Collapse
Affiliation(s)
- Xiaofan Lu
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yang Wang
- Department of Radiology, The Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Liyun Jiang
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jun Gao
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yue Zhu
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wenjun Hu
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jiashuo Wang
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xinjia Ruan
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhengbao Xu
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaowei Meng
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Bing Zhang
- Department of Radiology, The Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Fangrong Yan
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
15058
|
Han Y, Lu M, Zhou J. Buforin IIb induces androgen-independent prostate cancer cells apoptosis though p53 pathway in vitro. Toxicon 2019; 168:16-21. [PMID: 31229626 DOI: 10.1016/j.toxicon.2019.06.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 05/18/2019] [Accepted: 06/13/2019] [Indexed: 12/27/2022]
Abstract
Prostate cancer (PCa) is one of the most common male cancer types, androgen-independent prostate cancer possesses poor prognosis. Buforin IIb, an attractive antibacterial peptide derived from histone H2A, showed selective cytotoxicity against most cancer cell lines. However, the molecular mechanism of buforin IIb on prostate cancer cell has still not been determined. In this study, we found buforin IIb significantly inhibited the prostate cancer cells proliferation, Furthermore, buforin IIb-induced cell apoptosis through downregulation of pro-caspase 3/8/9, poly (ADP-ribose) polymerase PARP and anti-apoptotic Bcl-2 and upregulation of pro-apoptotic Bax. In addition, buforin IIb increased the expression of tumor suppressor p53 and its target genes - p21, fas, noxa and puma. The cytotoxicity of buforin IIb on PC-3 and Du-145 cells is decreased by p53 knockdown. In conclusion, our results indicated that buforin IIb induced PC-3 and Du-145 cell apoptosis and could be considered as a potential drug for prostate cancer.
Collapse
Affiliation(s)
- Yangyang Han
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China
| | - Ming Lu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China
| | - Jinsong Zhou
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China.
| |
Collapse
|
15059
|
Yuan Y, Gong H, Li Y, Zhang H, Li Y, Li W, Wang P, Shi R, Liu H, Chen J. [Effect of Apatinib on Invasion and Migration of Lung Cancer Cells
and Its Mechanism]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2019; 22:264-270. [PMID: 31109435 PMCID: PMC6533190 DOI: 10.3779/j.issn.1009-3419.2019.05.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
背景与目的 肺癌是世界上对人类健康产生重大危害的癌症之一,近年来靶向治疗的效果日益显著。阿帕替尼(Apatinib, YN968D1)是目前研究较热的多靶点抗肿瘤药物,本研究旨在探讨Apatinib对肺癌细胞生物学特性的影响和其可能的作用机制。 方法 体外培养肺腺癌细胞株H1299与H3255,CCK法、划痕实验及Transwell实验检测Apatinib对H1299与H3255细胞增殖、迁移及侵袭的影响,Western blot检测肿瘤血管生成和侵袭相关蛋白的表达。 结果 Apatinib显著抑制H1299与H3255的增殖、迁移及侵袭能力,并呈浓度依赖性。Western blot显示,随药物浓度增加,VEGF、VEGFR2、N-cadherin、MMP9、MMP2、Vimentin表达下调,E-cadherin表达上调。 结论 Apatinib可抑制肺腺癌细胞H1299、H3255的侵袭迁移能力,其机制可能通过调控上皮-间充质转化及基质金属蛋白酶相关蛋白的表达来实现。
Collapse
Affiliation(s)
- Yin Yuan
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hao Gong
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yongwen Li
- Tianjin Key Laboratory of lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hongbing Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ying Li
- Tianjin Key Laboratory of lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Weiting Li
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Pan Wang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ruifeng Shi
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hongyu Liu
- Tianjin Key Laboratory of lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jun Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Key Laboratory of lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
15060
|
Zhang Y, Xiong X, Huai Y, Dey A, Hossen NM, Roy RV, Elechalawar CK, Rao G, Bhattacharya R, Mukherjee P. Gold Nanoparticles Disrupt Tumor Microenvironment - Endothelial Cell Cross Talk To Inhibit Angiogenic Phenotypes in Vitro. Bioconjug Chem 2019; 30:1724-1733. [PMID: 31067032 PMCID: PMC6939887 DOI: 10.1021/acs.bioconjchem.9b00262] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is currently recognized that perpetual cross talk among key players in tumor microenvironment such as cancer cells (CCs), cancer associated fibroblasts (CAFs), and endothelial cells (ECs) plays a critical role in tumor progression, metastasis, and therapy resistance. Disruption of the cross talk may be useful to improve the outcome of therapeutics for which limited options are available. In the current study we investigate the use of gold nanoparticles (AuNPs) as a therapeutic tool to disrupt the multicellular cross talk within the TME cells with an emphasis on inhibiting angiogenesis. We demonstrate here that AuNPs disrupt signal transduction from TME cells (CCs, CAFs, and ECs) to ECs and inhibit angiogenic phenotypes in vitro. We show that conditioned media (CM) from ovarian CCs, CAFs, or ECs themselves induce tube formation and migration of ECs in vitro. Migration of ECs is also induced when ECs are cocultured with CCs, CAFs, or ECs. In contrast, CM from the cells treated with AuNPs or cocultured cells pretreated with AuNPs demonstrate diminished effects on ECs tube formation and migration. Mechanistically, AuNPs deplete ∼95% VEGF165 from VEGF single-protein solution and remove up to ∼45% of VEGF165 from CM, which is reflected on reduced activation of VEGF-Receptor 2 (VEGFR2) as compared to control CM. These results demonstrate that AuNPs inhibit angiogenesis via blockade of VEGF-VEGFR2 signaling from TME cells to endothelial cells.
Collapse
Affiliation(s)
- Yushan Zhang
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Xunhao Xiong
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Yanyan Huai
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Anindya Dey
- Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Nazir Md Hossen
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Ram Vinod Roy
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Chandra Kumar Elechalawar
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Geeta Rao
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Priyabrata Mukherjee
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| |
Collapse
|
15061
|
FBXO22 mediates polyubiquitination and inactivation of LKB1 to promote lung cancer cell growth. Cell Death Dis 2019; 10:486. [PMID: 31217475 PMCID: PMC6584689 DOI: 10.1038/s41419-019-1732-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 05/19/2019] [Accepted: 05/27/2019] [Indexed: 12/19/2022]
Abstract
Liver kinase B1 (LKB1) regulates both cell growth and energy metabolism. Inactivated mutations of LKB1, observed in 20–30% of nonsmall cell lung cancers (NSCLC), contribute significantly to lung cancer malignancy progression. However, the upstream signalings regulating LKB1 activity remain incompletely understood. Here, we present evidence that FBXO22 interacts with and promotes polyubiquitination of LKB1. More intriguingly, FBXO22 mediates Lys-63-linked LKB1 polyubiquitination and inhibits kinase activity of LKB1. Furthermore, over-expression of FBXO22 promotes NSCLC cell growth through inhibiting LKB1-AMPK-mTOR signaling in vitro and in vivo. Clinically, FBXO22 is highly expressed in human lung adenocarcinoma and high FBXO22 expression predicts significant poor prognosis. Our study provides new insights into the upstream regulation of LKB1 activation and identifies FBXO22 as a potential therapeutic target for lung cancer treatment.
Collapse
|
15062
|
Demirci S, Hayal TB, Kıratlı B, Şişli HB, Demirci S, Şahin F, Doğan A. Design and synthesis of phenylpiperazine derivatives as potent anticancer agents for prostate cancer. Chem Biol Drug Des 2019; 94:1584-1595. [PMID: 31148379 DOI: 10.1111/cbdd.13575] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/23/2019] [Accepted: 05/26/2019] [Indexed: 12/19/2022]
Abstract
Novel thiourea (5a, 5b) and thiazolidinone derivatives (6a, 6b) were synthesized by hybridizing molecules starting from the compound 6-(4-phenylpiperazin-1-yl)pyridin-3-amine (4) which is known to show anticancer activity. The synthesis of the leading compound was carried out by using 1-(5-nitropyridin-2-yl)-4-phenylpiperazine (3) which was obtained by a novel method of the reaction of 2-chloro-5-nitropyridine (1) and N-phenylpiperazine (2). The structures of the compounds were confirmed using FTIR, 1 H NMR, 13 C NMR, HRMS spectroscopic methods and elemental analysis. The organic molecules were tested for their anticancer activities against prostate cancer (PC) cell lines: DU 145, PC-3 and LNCaP. As the compound 5a exerted the highest cytotoxic activity, IC50 concentrations of compound 5a were further investigated in terms of morphology, colony-forming ability, RNA expression, fragmented DNA and cell cycle distributions of PC cell lines. Overall data revealed that compound 5a treatment induces apoptosis and DNA fragmentation in PC cell lines and inhibits cell cycle progression resulting in the accumulation of cells in either the G1 or the S phases.
Collapse
Affiliation(s)
- Serpil Demirci
- Department of Medical Services and Techniques, Vocational High School of Health Services, Giresun University, Giresun, Turkey
| | - Taha Bartu Hayal
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Binnur Kıratlı
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Hatice Burcu Şişli
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Selami Demirci
- Cellular and Molecular Therapeutics, Sickle Cell Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Ayşegül Doğan
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| |
Collapse
|
15063
|
Myoferlin Contributes to the Metastatic Phenotype of Pancreatic Cancer Cells by Enhancing Their Migratory Capacity through the Control of Oxidative Phosphorylation. Cancers (Basel) 2019; 11:cancers11060853. [PMID: 31248212 PMCID: PMC6628295 DOI: 10.3390/cancers11060853] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/13/2019] [Accepted: 06/16/2019] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies with an overall survival of 5% and is the second cause of death by cancer, mainly linked to its high metastatic aggressiveness. Accordingly, understanding the mechanisms sustaining the PDAC metastatic phenotype remains a priority. In this study, we generated and used a murine in vivo model to select clones from the human Panc-1 PDAC cell line that exhibit a high propensity to seed and metastasize into the liver. We showed that myoferlin, a protein previously reported to be overexpressed in PDAC, is significantly involved in the migratory abilities of the selected cells. We first report that highly metastatic Panc-1 clones expressed a significantly higher myoferlin level than the corresponding low metastatic ones. Using scratch wound and Boyden’s chamber assays, we show that cells expressing a high myoferlin level have higher migratory potential than cells characterized by a low myoferlin abundance. Moreover, we demonstrate that myoferlin silencing leads to a migration decrease associated with a reduction of mitochondrial respiration. Since mitochondrial oxidative phosphorylation has been shown to be implicated in the tumor progression and dissemination, our data identify myoferlin as a valid potential therapeutic target in PDAC.
Collapse
|
15064
|
Sun R, Meng X, Wang W, Liu B, Lv X, Yuan J, Zeng L, Chen Y, Yuan B, Yang S. Five genes may predict metastasis in non-small cell lung cancer using bioinformatics analysis. Oncol Lett 2019; 18:1723-1732. [PMID: 31423239 PMCID: PMC6607402 DOI: 10.3892/ol.2019.10498] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 05/14/2019] [Indexed: 12/16/2022] Open
Abstract
Lung cancer is one of the most common types of malignancy worldwide. The prognosis of lung cancer is poor, due to the onset of metastases. The aim of the present study was to examine lung cancer metastasis-associated genes. To identify novel metastasis-associated targets, our previous study detected the differentially expressed mRNAs and long non-coding RNAs between the large-cell lung cancer high-metastatic 95D cell line and the low-metastatic 95C cell line by microarray assay. In the present study, these differentially expressed genes (DEGs) were analyzed via bioinformatics methods, including Gene Ontology functional analysis and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis. A protein-protein interaction network was subsequently constructed using the Search Tool for the Retrieval of Interacting Genes/Proteins online database and Cytoscape software, and 17 hub genes were screened out on the basis of connectivity degree. These hub genes were further validated in lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) using the online Gene Expression Profiling Interactive Analysis database. A total of seven hub genes were identified to be significantly differentially expressed in LUAD and LUSC. The prognostic information was detected using Kaplan-Meier plotter. As a result, five genes were revealed to be closely associated with the overall survival time of patients with lung cancer, including phosphoinositide-3-kinase regulatory subunit 1, FYN, thrombospondin 1, nonerythrocytic α-spectrin 1 and secreted phosphoprotein 1. In addition, lung cancer and adjacent lung tissue samples were used to validate these hub genes by reverse transcription-quantitative polymerase chain reaction. In conclusion, the results of the present study may provide novel metastasis-associated therapeutic strategies or potential biomarkers in non-small cell lung cancer.
Collapse
Affiliation(s)
- Ruiying Sun
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xia Meng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Wei Wang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Boxuan Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xin Lv
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jingyan Yuan
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Lizhong Zeng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yang Chen
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Bo Yuan
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Shuanying Yang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
15065
|
Moreira VM, Almeida D, da Silva Franco CC, Gomes RM, Palma-Rigo K, Prates KV, Tófolo LP, Malta A, Francisco FA, Pavanello A, Previate C, da Silva Silveira S, Ribeiro TA, Martins IP, de Moraes AMP, Matiusso CCI, Saavedra LPJ, de Barros Machado KG, Fabbri Corá T, Gongora A, Cardozo LE, da Silva PHO, Venci R, Vieira E, de Oliveira JC, Miranda RA, de Souza HM, Miksza D, da Costa Lima LD, de Castro-Prado MAA, Rinaldi W, de Freitas Mathias PC. Moderate exercise training since adolescence reduces Walker 256 tumour growth in adult rats. J Physiol 2019; 597:3905-3925. [PMID: 31210356 DOI: 10.1113/jp277645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 05/07/2019] [Indexed: 12/19/2022] Open
Abstract
KEY POINTS Cancer growth, cell proliferation and cachexia index can be attenuated by the beneficial programming effect of moderate exercise training, especially if it begins in adolescence. Walker 256 tumour-bearing rats who started exercise training during adolescence did not revert the basal low glycaemia and insulinaemia observed before tumour cell inoculation. The moderate exercise training improved glucose tolerance and peripheral insulin sensitivity only in rats exercised early in adolescence. The chronic effects of our exercise protocol are be beneficial to prevent cancer cachexia and hold clear potential as a nonpharmacological therapy of insulin sensitization. ABSTRACT We tested the hypothesis that moderate exercise training, performed early, starting during adolescence or later in life during adulthood, can inhibit tumour cell growth as a result of changes in biometric and metabolic markers. Male rats that were 30 and 70 days old performed a treadmill running protocol over 8 weeks for 3 days week-1 , 44 min day-1 and at 55-65% V ̇ O 2 max . After the end of training, a batch of rats was inoculated with Walker 256 carcinoma cells. At 15 days after carcinoma cell inoculation, the tumour was weighed and certain metabolic parameters were evaluated. The data demonstrated that physical performance was better in rats that started exercise training during adolescence according to the final workload and V ̇ O 2 max . Early or later moderate exercise training decreased the cachexia index, cell proliferation and tumour growth; however, the effects were more pronounced in rats that exercised during adolescence. Low glycaemia, insulinaemia and tissue insulin sensitivity was not reverted in Walker 256 tumour-bearing rats who trained during adolescence. Cancer growth can be attenuated by the beneficial programming effect of moderate exercise training, especially if it begins during adolescence. In addition, improvement in glucose-insulin homeostasis might be involved in this process.
Collapse
Affiliation(s)
- Veridiana Mota Moreira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil.,Department of Physical Education, State University of Maringá, Maringá, PR, Brazil
| | - Douglas Almeida
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | | | | | - Kesia Palma-Rigo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Kelly Valério Prates
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Laize Peron Tófolo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil.,Department of Physical Education, State University of Maringá, Maringá, PR, Brazil
| | - Ananda Malta
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Flávio Andrade Francisco
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Audrei Pavanello
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Carina Previate
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Sandra da Silva Silveira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Tatiane Aparecida Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Isabela Peixoto Martins
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Ana Maria Praxedes de Moraes
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Camila Cristina Ianoni Matiusso
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Lucas Paulo Jacinto Saavedra
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Katia Gama de Barros Machado
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Thauany Fabbri Corá
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Adriane Gongora
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Lucas Eduardo Cardozo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil.,Department of Physical Education, State University of Maringá, Maringá, PR, Brazil
| | - Paulo Henrique Olivieri da Silva
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil.,Department of Physical Education, State University of Maringá, Maringá, PR, Brazil
| | - Renan Venci
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Elaine Vieira
- Post-Graduate Program of Physical Education, Catholic University of Brasília, Águas Claras, DF, Brazil
| | | | - Rosiane Aparecida Miranda
- Laboratory of Molecular Endocrinology, Carlos Chagas Filho Biophysis Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Daniele Miksza
- Department of Physiology, State University of Londrina, Londrina, PR, Brazil
| | - Luiz Delmar da Costa Lima
- Superior School of Physical Education and Physical Therapy of Goiás State, State University of Goiás, Goiânia, GO, Brazil
| | - Marialba Avezum Alves de Castro-Prado
- Laboratory of Microorganisms Genetics and Mutagenesis, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Wilson Rinaldi
- Department of Physical Education, State University of Maringá, Maringá, PR, Brazil
| | - Paulo Cezar de Freitas Mathias
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| |
Collapse
|
15066
|
Bam R, Laffey M, Nottberg K, Lown PS, Hackel BJ, Wilson KE. Affibody-Indocyanine Green Based Contrast Agent for Photoacoustic and Fluorescence Molecular Imaging of B7-H3 Expression in Breast Cancer. Bioconjug Chem 2019; 30:1677-1689. [PMID: 31082216 PMCID: PMC6745046 DOI: 10.1021/acs.bioconjchem.9b00239] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Spectroscopic photoacoustic (sPA) molecular imaging has high potential for identification of exogenous contrast agents targeted to specific markers. Antibody-dye conjugates have recently been used extensively for preclinical sPA and other optical imaging modalities for highly specific molecular imaging of breast cancer. However, antibody-based agents suffer from long circulation times that limit image specificity. Here, the efficacy of a small protein scaffold, the affibody (ABY), conjugated to indocyanine green (ICG), a near-infrared fluorescence dye, as a targeted molecular imaging probe is demonstrated. In particular, B7-H3 (CD276), a cellular receptor expressed in breast cancer, was imaged via sPA and fluorescence molecular imaging to differentiate invasive tumors from normal glands in mice. Administration of ICG conjugated to an ABY specific to B7-H3 (ABYB7-H3-ICG) showed significantly higher signal in mammary tumors compared to normal glands of mice. ABYB7-H3-ICG is a compelling scaffold for molecular sPA imaging for breast cancer detection.
Collapse
Affiliation(s)
- Rakesh Bam
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| | - Makenna Laffey
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| | - Katharine Nottberg
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| | - Patrick S. Lown
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Benjamin J. Hackel
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Katheryne E. Wilson
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
15067
|
Tewari KS, Burger RA, Enserro D, Norquist BM, Swisher EM, Brady MF, Bookman MA, Fleming GF, Huang H, Homesley HD, Fowler JM, Greer BE, Boente M, Liang SX, Ye C, Bais C, Randall LM, Chan JK, Ferriss JS, Coleman RL, Aghajanian C, Herzog TJ, DiSaia PJ, Copeland LJ, Mannel RS, Birrer MJ, Monk BJ. Final Overall Survival of a Randomized Trial of Bevacizumab for Primary Treatment of Ovarian Cancer. J Clin Oncol 2019; 37:2317-2328. [PMID: 31216226 DOI: 10.1200/jco.19.01009] [Citation(s) in RCA: 306] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
PURPOSE We report the final, protocol-specified analysis of overall survival (OS) in GOG-0218, a phase III, randomized trial of bevacizumab in women with newly diagnosed ovarian, fallopian tube, or primary peritoneal carcinoma. METHODS A total of 1,873 women with incompletely resected stage III to IV disease were randomly assigned 1:1:1 to six 21-day cycles of intravenous carboplatin (area under the concentration v time curve 6) and paclitaxel (175 mg/m2) versus chemotherapy plus concurrent bevacizumab (15 mg/kg, cycles 2 to 6) versus chemotherapy plus concurrent and maintenance bevacizumab (cycles 2 to 22). Inclusion criteria included a Gynecologic Oncology Group performance status of 0 to 2 and no history of clinically significant vascular events or evidence of intestinal obstruction. OS was analyzed in the intention-to-treat population. A total of 1,195 serum and/or tumor specimens were sequenced for BRCA1/2 and damaging mutations in homologous recombination repair (HRR) genes. Intratumoral microvessel density was studied using CD31 immunohistochemistry. RESULTS Median follow-up was 102.9 months. Relative to control (n = 625), for patients receiving bevacizumab-concurrent (n = 625), the hazard ratio (HR) of death was 1.06 (95% CI, 0.94 to 1.20); for bevacizumab-concurrent plus maintenance (n = 623), the HR was 0.96 (95% CI, 0.85 to 1.09). Disease-specific survival was not improved in any arm. No survival advantage was observed after censoring patients who received bevacizumab at crossover or as second line. Median OS for stage IV bevacizumab-concurrent plus maintenance was 42.8 v 32.6 months for stage IV control (HR, 0.75; 95% CI, 0.59 to 0.95). Relative to wild type, the HR for death for BRCA1/2 mutated carcinomas was 0.62 (95% CI, 0.52 to 0.73), and for non-BRCA1/2 HRR, the HR was 0.65 (95% CI, 0.51 to 0.85). BRCA1/2, HRR, and CD31 were not predictive of bevacizumab activity. CONCLUSION No survival differences were observed for patients who received bevacizumab compared with chemotherapy alone. Testing for BRCA1/2 mutations and homologous recombination deficiency is essential.
Collapse
Affiliation(s)
| | - Robert A Burger
- University of Pennsylvania Abramson Cancer Center, Philadelphia, PA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - John K Chan
- California Pacific Medical Center Research Institute, San Francisco, CA
| | - J Stuart Ferriss
- Dell Seton Medical Center at The University of Texas, Austin, TX
| | | | | | | | - Philip J DiSaia
- University of California, Irvine, Medical Center, Orange, CA
| | | | - Robert S Mannel
- University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | | | - Bradley J Monk
- University of Arizona and Creighton University, Phoenix, AZ
| |
Collapse
|
15068
|
Jammal N, Pan E, Hurwitz M, Abramovitz RB. Outcomes of combination therapy with tyrosine kinase inhibitors and immune checkpoint inhibitors in metastatic renal cell carcinoma – A retrospective study. J Oncol Pharm Pract 2019; 26:556-563. [DOI: 10.1177/1078155219854797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction Renal cell carcinoma (RCC) is a highly vascularized and immunogenic tumor. At the time of this study, there was limited published data on the combination of tyrosine kinase inhibitors and immune checkpoint inhibitors in patients who were heavily pretreated. At our institution, providers have used these combinations in heavily pretreated patients. Methods We conducted a retrospective review of patients receiving this combination with the primary objectives of assessing duration of therapy and toxicities and a secondary objective of disease progression at six months. We included adult patients with confirmed mRCC receiving combination therapy (immune checkpoint inhibitors/tyrosine kinase inhibitors) any time after January 2015. Electronic medical records were reviewed for pertinent data and follow-up descriptive statistics were performed. Results Fifteen patients were on combination immune checkpoint inhibitors/tyrosine kinase inhibitors, with a median of three lines of previous therapy. The median duration of combination therapy was 7.2 months (range: 0.2 to 39.8) with 126 incidences of toxicities. The most frequent toxicity was fatigue (n = 15), followed by diarrhea (n = 8), anorexia (n = 7) and palmar-plantar erythrodysesthesia (n = 7). Overall, 9 (60%) patients experienced at least one grade 3 or 4 toxicity. Eight of 15 (53%) patients remained on therapy at the six-month mark and did not have progression confirmed by an oncologist. Of the 15 patients, 10 discontinued therapy due to progression, 2 due to intolerable side effects, 2 transitioned to end of life care, and 1 patient was still ongoing at the time of data collection. Conclusion Based on this review, it appears that combination tyrosine kinase inhibitors/immune checkpoint inhibitors therapy in pre-treated patients with mRCC is tolerable and beneficial.
Collapse
Affiliation(s)
| | - Eva Pan
- Yale-New Haven Hospital, New Haven, CT, USA
| | | | | |
Collapse
|
15069
|
Khatri G, Pedrosa I, Ananthakrishnan L, Leon AD, Fetzer DT, Leyendecker J, Singal AG, Xi Y, Yopp A, Yokoo T. Abbreviated‐protocol screening MRI vs. complete‐protocol diagnostic MRI for detection of hepatocellular carcinoma in patients with cirrhosis: An equivalence study using LI‐RADS v2018. J Magn Reson Imaging 2019; 51:415-425. [DOI: 10.1002/jmri.26835] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 05/17/2019] [Accepted: 05/21/2019] [Indexed: 12/25/2022] Open
Affiliation(s)
- Gaurav Khatri
- Department of RadiologyUniversity of Texas Southwestern Medical Center Dallas Texas
| | - Ivan Pedrosa
- Department of RadiologyUniversity of Texas Southwestern Medical Center Dallas Texas
| | | | - Alberto Diaz Leon
- Department of RadiologyUniversity of Texas Southwestern Medical Center Dallas Texas
| | - David T. Fetzer
- Department of RadiologyUniversity of Texas Southwestern Medical Center Dallas Texas
| | - John Leyendecker
- Department of RadiologyUniversity of Texas Southwestern Medical Center Dallas Texas
| | - Amit G. Singal
- Department of Internal MedicineUniversity of Texas Southwestern Medical Center Dallas Texas
| | - Yin Xi
- Department of RadiologyUniversity of Texas Southwestern Medical Center Dallas Texas
| | - Adam Yopp
- Department of SurgeryUniversity of Texas Southwestern Medical Center Dallas Texas
| | - Takeshi Yokoo
- Department of RadiologyUniversity of Texas Southwestern Medical Center Dallas Texas
| |
Collapse
|
15070
|
Yang H, Fu G, Liu F, Hu C, Lin J, Tan Z, Fu Y, Ji F, Cao M. LncRNA THOR promotes tongue squamous cell carcinomas by stabilizing IGF2BP1 downstream targets. Biochimie 2019; 165:9-18. [PMID: 31220513 DOI: 10.1016/j.biochi.2019.06.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 06/14/2019] [Indexed: 02/07/2023]
Abstract
THOR, a highly conserved lncRNA, is potentially involved in various cancer development. However, its involvement in tongue squamous cell carcinoma (TSCC) remains unclear. The present study aims to explore the biological function and molecular mechanism of THOR in TSCC progression. The expressions of THOR and IGF2BP1 in TSCC tissues and adjacent non-cancerous tongue tissues (ANT) were examined through qRT-PCR. THOR levels were manipulated in TSCC cells to explore its function in cancer progression in vitro and in vivo, which were subsequently evaluated by CCK8, colony formation assay, flow cytometry, xenograft tumor assays. In situ hybridization, RIP and Western blot assay were performed to explore the underlying molecular mechanisms. We discovered that THOR and IGF2BP1 were dramatically upregulated in TSCC tissues. The expression of THOR is positively correlated with IGF2BP1 mRNA level. THOR mediated IGF2 expression via interacting with IGF2BP1, and affected the downstream MEK-ERK signaling pathway to regulate TSCC cells proliferation. THOR/IGF2BP1/IGF2-MEK-ERK axis regulated the proliferation of TSCC cells, implying that THOR would be a promising therapeutic target for TSCC patients.
Collapse
Affiliation(s)
- Haojie Yang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ganglan Fu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Funing Liu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chuwen Hu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Junjie Lin
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zicong Tan
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yanni Fu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fengtao Ji
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Minghui Cao
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
15071
|
Duration of Adjuvant Chemotherapy in Colon Cancer: Current Standards and New Updates. CURRENT COLORECTAL CANCER REPORTS 2019. [DOI: 10.1007/s11888-019-00438-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
15072
|
Spartalis C, Schmidt EM, Elmasry M, Schulz GB, Kirchner T, Horst D. In vivo effects of chemotherapy on oncogenic pathways in colorectal cancer. Cancer Sci 2019; 110:2529-2539. [PMID: 31119819 PMCID: PMC6676136 DOI: 10.1111/cas.14077] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/06/2019] [Accepted: 05/20/2019] [Indexed: 12/22/2022] Open
Abstract
Patients with advanced colorectal cancer often are treated with systemic cytotoxic therapy using fluorouracil (5‐FU), oxaliplatin, irinotecan, and FOLFOX or FOLFIRI combination protocols. Additionally, signaling pathways that are active in colorectal cancer can be therapeutically targeted. Herein, we examined whether chemotherapy impacts on WNT, MAPK and NOTCH signaling pathways in xenograft models of colon cancer. Furthermore, we tested whether combining chemotherapy with MAPK and NOTCH inhibition has superior therapeutic effects. We show that colon cancer cells with high WNT, MAPK and NOTCH activity are variably affected but generally persist in xenograft tumors under different chemotherapeutic regimens, indicating limited effects of cytotoxic therapy on oncogenic signaling pathways. Although these results provided a rationale to additionally target pathway activity, we found no significant increase in therapy response when combining MAPK and NOTCH inhibition with fluorouracil chemotherapy. We attribute this finding to a decrease in tumor cell proliferation upon MAPK and NOTCH inhibition, resulting in reduced effectiveness of cytotoxic treatment. Therapeutic benefits of combining chemotherapy with targeting of oncogenic signaling pathways must therefore be critically evaluated for patients with colorectal cancer.
Collapse
Affiliation(s)
- Christoph Spartalis
- Institute of Pathology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Eva Marina Schmidt
- Institute of Pathology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Manal Elmasry
- Institute of Pathology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | | | - Thomas Kirchner
- Institute of Pathology, Ludwig-Maximilians-Universität Munich, Munich, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David Horst
- Institute of Pathology, Ludwig-Maximilians-Universität Munich, Munich, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
15073
|
Abaricia S, Van Tine BA. Management of localized extremity and retroperitoneal soft tissue sarcoma. Curr Probl Cancer 2019; 43:273-282. [PMID: 31221500 DOI: 10.1016/j.currproblcancer.2019.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 06/08/2019] [Indexed: 11/16/2022]
Abstract
The optimal management of localized soft tissue sarcomas of the extremities and retroperitoneum involves a high volume multidisciplinary team with expertise in sarcoma. In this review, we will highlight the importance of the sarcoma pathologist and imaging techniques prior to surgery and radiation. In addition, the data on neoadjuvant and adjuvant chemotherapy will be discussed. Finally, consideration is given to the importance of identifying genetic cancer predispositions, multidisciplinary management, long-term survivorship, and the current clinical trials for patients undergoing curative intent management.
Collapse
Affiliation(s)
- Sarah Abaricia
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Brian Andrew Van Tine
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri; Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
15074
|
Ovarian Cancer Immunotherapy: Turning up the Heat. Int J Mol Sci 2019; 20:ijms20122927. [PMID: 31208030 PMCID: PMC6628106 DOI: 10.3390/ijms20122927] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 12/20/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the leading cause of death among gynecological malignancies. Despite surgery and chemotherapy, 5-years survival rates have improved only modestly over the past few decades remaining at 45% for advanced stages. Therefore, novel therapies are urgently needed. The presence of tumor-infiltrating lymphocytes (TILs) in OC tumor microenvironment (TME) has already proved to be correlated with overall survival (OS), while immune evasion mechanisms are associated with poor prognosis. Although these data indicate that immunotherapy has a strong rationale in OC, single agent immune-checkpoints inhibitors (ICIs) have shown only modest results in this malignancy. In this review, we will discuss immune-targeting combination therapies and adoptive cell therapy (ACT), highlighting the challenges represented by these strategies, which aim at disrupting the stroma-tumor barrier to boost immune system against ovarian cancer.
Collapse
|
15075
|
Chromogranin-A Expression as a Novel Biomarker for Early Diagnosis of Colon Cancer Patients. Int J Mol Sci 2019; 20:ijms20122919. [PMID: 31207989 PMCID: PMC6628020 DOI: 10.3390/ijms20122919] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/06/2019] [Accepted: 06/12/2019] [Indexed: 12/24/2022] Open
Abstract
Colon cancer is one of the major causes of cancer death worldwide. The five-year survival rate for the early-stage patients is more than 90%, and only around 10% for the later stages. Moreover, half of the colon cancer patients have been clinically diagnosed at the later stages. It is; therefore, of importance to enhance the ability for the early diagnosis of colon cancer. Taking advantages from our previous studies, there are several potential biomarkers which have been associated with the early diagnosis of the colon cancer. In order to investigate these early diagnostic biomarkers for colon cancer, human chromogranin-A (CHGA) was further analyzed among the most powerful diagnostic biomarkers. In this study, we used a logistic regression-based meta-analysis to clarify associations of CHGA expression with colon cancer diagnosis. Both healthy populations and the normal mucosa from the colon cancer patients were selected as the double normal controls. The results showed decreased expression of CHGA in the early stages of colon cancer as compared to the normal controls. The decline of CHGA expression in the early stages of colon cancer is probably a new diagnostic biomarker for colon cancer diagnosis with high predicting possibility and verification performance. We have also compared the diagnostic powers of CHGA expression with the typical oncogene KRAS, classic tumor suppressor TP53, and well-known cellular proliferation index MKI67, and the CHGA showed stronger ability to predict early diagnosis for colon cancer than these other cancer biomarkers. In the protein-protein interaction (PPI) network, CHGA was revealed to share some common pathways with KRAS and TP53. CHGA might be considered as a novel, promising, and powerful biomarker for early diagnosis of colon cancer.
Collapse
|
15076
|
Alwhaibi A, Verma A, Artham S, Adil MS, Somanath PR. Nodal pathway activation due to Akt1 suppression is a molecular switch for prostate cancer cell epithelial-to-mesenchymal transition and metastasis. Biochem Pharmacol 2019; 168:1-13. [PMID: 31202735 DOI: 10.1016/j.bcp.2019.06.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/10/2019] [Indexed: 12/15/2022]
Abstract
Several studies have unraveled the negative role of Akt1 in advanced cancers, including metastatic prostate cancer (mPCa). Hence, understanding the consequences of targeting Akt1 in the mPCa and identifying its downstream novel targets is essential. We studied how Akt1 deletion in PC3 and DU145 cells activates the Nodal pathway and promotes PCa epithelial-to-mesenchymal transition (EMT) and metastasis. Here we show that Akt1 loss increases Nodal expression in PCa cells accompanied by activation of FoxO1/3a, and EMT markers Snail and N-cadherin as well as loss of epithelial marker E-cadherin. Treatment with FoxO inhibitor AS1842856 abrogated the Nodal expression in Akt1 deleted PCa cells. Akt1 deficient PCa cells exhibited enhanced cell migration and invasion in vitro and lung metastasis in vivo, which were attenuated by treatment with Nodal pathway inhibitor SB505124. Interestingly, Nodal mRNA analysis from two genomic studies in cBioportal showed a positive correlation between Nodal expression and Gleason score indicating the positive role of Nodal in human mPCa. Collectively, our data demonstrate Akt1-FoxO3a-Nodal pathway as an important mediator of PCa metastasis and present Nodal as a potential target to treat mPCa patients.
Collapse
Affiliation(s)
- Abdulrahman Alwhaibi
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Arti Verma
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Sandeep Artham
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Mir S Adil
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States; Department of Medicine, Vascular Biology Center and Cancer Center, Augusta University, Augusta, GA 30912, United States.
| |
Collapse
|
15077
|
Hanna KS. Updates in the management of chronic lymphocytic leukemia/small lymphocytic leukemia. J Oncol Pharm Pract 2019; 26:146-155. [PMID: 31189420 DOI: 10.1177/1078155219853030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Chronic lymphocytic leukemia is one of the most common lymphoid malignancies. Often treatment modalities are tailored to individual patients based on age, presence of comorbidities and cytogenetics. The advent of ibrutinib has significantly changed the management of the disease in all patient groups and has had the largest impact on clinical practice to date. Over the last 15 years, a series of trials have established that chemoimmunotherapy improves both progression-free survival and overall survival compared to chemotherapy alone. Despite its proven role, efficacy of ibrutinib has not been well established in young, fit patients and in comparison with standard care and as combination therapy with other agents such as venetoclax. New data have strengthened the role of ibrutinib in the front-line setting and establish its place in therapy. In addition, combination therapies are geared to achieve negative minimal residual disease and allow patients to potentially be off of therapy. The management of this leukemia has extensively changed over the past years, and this review article will aim to highlight key trials that have changed practice and led to guideline updates. It is not unlikely that treatment modalities will continue to improve in light of new data.
Collapse
Affiliation(s)
- Kirollos S Hanna
- Mayo Clinic College of Medicine, Mayo Clinic & University of Minnesota Medical Center, Rochester, MN, USA
| |
Collapse
|
15078
|
Liu Y, Zhang YM, Ma FB, Pan SR, Liu BZ. Long noncoding RNA HOXA11-AS promotes gastric cancer cell proliferation and invasion via SRSF1 and functions as a biomarker in gastric cancer. World J Gastroenterol 2019; 25:2763-2775. [PMID: 31235999 PMCID: PMC6580350 DOI: 10.3748/wjg.v25.i22.2763] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/15/2019] [Accepted: 05/03/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is the fourth most frequent malignancy all over the world. The diagnosis of GC is challenging and the prognosis of GC is very unfavorable. Accumulating evidence reveals that serum long noncoding RNAs (lncRNAs) can function as biomarkers in various types of cancers, including GC.
AIM To explore the level and molecular mechanism of the lncRNA HOXA11-AS in GC and the diagnostic and prognostic significance of serum HOXA11-AS in GC.
METHODS HOXA11-AS levels in GC tissue, cell lines, and serum samples were measured. The correlation between HOXA11-AS expression and clinicopathological characteristics was analyzed. The role of HOXA11-AS in the diagnosis and prognosis of GC was evaluated. Cell function assays were performed for exploration of the roles of HOXA11-AS in GC cells. Moreover, Western blot was performed to explore the target regulated by HOXA11-AS in GC cells.
RESULTS Up-regulation of HOXA11-AS was found in GC tissues, cell lines, and serum samples. In GC patients, decreased serum HOXA11-AS levels were negatively related with tumor size, TNM stage, and lymph node metastasis. The area under the receiver operating characteristic curve of serum HOXA11-AS in the diagnosis of GC was 0.924 (95%CI: 0.881-0.967; sensitivity, 0.787; specificity 0.978). Results of the Kaplan-Meier survival curves suggested the GC patients with a lower HOXA11-AS level having a better overall survival rate. HOXA11-AS promoted GC cell proliferation and invasion. SRSF1 may be the target regulated by HOXA11-AS in GC cells.
CONCLUSION HOXA11-AS promotes GC cell proliferation and invasion via SRSF1 and may function as a promising marker in GC.
Collapse
Affiliation(s)
- Yun Liu
- Department of Operating Room, Binzhou People's Hospital, Binzhou 256610, Shandong Province, China
| | - Yu-Mei Zhang
- Department of Return Visit, Binzhou People's Hospital, Binzhou 256610, Shandong Province, China
| | - Feng-Bo Ma
- Department of Gastroenterology, Binzhou People's Hospital, Binzhou 256610, Shandong Province, China
| | - Su-Rong Pan
- Department of Gastroenterology, Binzhou People's Hospital, Binzhou 256610, Shandong Province, China
| | - Bao-Zhen Liu
- Department of Gastroenterology, Binzhou People's Hospital, Binzhou 256610, Shandong Province, China
| |
Collapse
|
15079
|
Neoantigens Derived from Recurrently Mutated Genes as Potential Immunotherapy Targets for Gastric Cancer. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8103142. [PMID: 31312661 PMCID: PMC6595338 DOI: 10.1155/2019/8103142] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/12/2019] [Indexed: 12/30/2022]
Abstract
Neoantigens are optimal tumor-specific targets for T-cell based immunotherapy, especially for patients with “undruggable” mutated driver genes. T-cell immunotherapy can be a “universal” treatment for HLA genotype patients sharing same oncogenic mutations. To identify potential neoantigens for therapy in gastric cancer, 32 gastric cancer patients were enrolled in our study. Whole exome sequencing data from these patients was processed by TSNAD software to detect cancer somatic mutations and predict neoantigens. The somatic mutations between different patients suggested a high interpatient heterogeneity. C>A and C>T substitutions are common, suggesting an active nucleotide excision repair. The number of predicted neoantigens was significantly higher in patients at stage T1a compared to in patients at T2 or T4b. Six genes (PIK3CA, FAT4, BRCA2, GNAQ, LRP1B, and PREX2) were found as recurrently mutated driver genes in our study. Combining with highly frequent HLA alleles, several neoantigens derived from six recurrently mutated genes were considered as potential targets for further immunotherapy.
Collapse
|
15080
|
Song YP, McWilliam A, Hoskin PJ, Choudhury A. Organ preservation in bladder cancer: an opportunity for truly personalized treatment. Nat Rev Urol 2019; 16:511-522. [PMID: 31197260 DOI: 10.1038/s41585-019-0199-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2019] [Indexed: 02/07/2023]
Abstract
Radical treatment of many solid tumours has moved from surgery to multimodal organ preservation strategies combining systemic and local treatments. Trimodality bladder-preserving treatment (TMT) comprises maximal transurethral resection of the bladder tumour followed by radiotherapy and concurrent radiosensitizing treatment, thereby sparing the urinary bladder. From the patient's perspective, the choice of maintaining quality of life without a negative effect on the chances of cure and long-term survival is attractive. In muscle-invasive bladder cancer (MIBC), the evidence shows comparable clinical outcomes between patients undergoing radical cystectomy and TMT. Despite this evidence, many patients continue to be offered radical surgery as the standard-of-care treatment. Improvements in radiotherapy techniques with adaptive radiotherapy and advances in imaging translate to increases in the accuracy of treatment delivery and reductions in long-term toxicities. With the advent of novel biomarkers promising improved prediction of treatment response, stratification of patients for different treatments on the basis of tumour biology could soon be a reality. The future of oncological treatment lies in personalized medicine with the combination of technological and biological advances leading to truly bespoke management for patients with MIBC.
Collapse
Affiliation(s)
- Yee Pei Song
- Manchester Cancer Research Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK. .,Department of Clinical Oncology, The Christie Hospital NHS Foundation Trust, Manchester, UK.
| | - Alan McWilliam
- Manchester Cancer Research Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK.,Department of Clinical Oncology, The Christie Hospital NHS Foundation Trust, Manchester, UK
| | - Peter J Hoskin
- Manchester Cancer Research Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK.,Mount Vernon Cancer Centre, Northwood, Middlesex, United Kingdom
| | - Ananya Choudhury
- Manchester Cancer Research Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK.,Department of Clinical Oncology, The Christie Hospital NHS Foundation Trust, Manchester, UK
| |
Collapse
|
15081
|
Xu B, Lv W, Li X, Zhang L, Lin J. Clinicopathological significance of TM4SF5 expression in human hepatocellular carcinoma tissues. Oncol Lett 2019; 17:5187-5192. [PMID: 31186734 DOI: 10.3892/ol.2019.10210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 02/22/2019] [Indexed: 01/25/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-related mortality. Recent studies have indicated that transmembrane 4 L six family member 5 (TM4SF5) serves a vital role in tumor progression and metastasis in human cell lines and mouse models. However, little is known about the association between TM4SF5 expression and clinicopathological factors. The aim of the current study was to investigate this association and evaluate overall survival (OS) of patients. Immunohistochemistry and clinical record analysis revealed that TM4SF5 expression was significantly downregulated in HCC tissues. In addition, TM4SF5 expression was significantly associated with tumor size, vascular invasion, tumor differentiation, and tumor-node-metastasis stage. The survival analysis also demonstrated that low TM4SF5 expression resulted in shorter OS. In conclusion, the association between TM4SF5 expression and clinicopathologic factors was established, and prognostic significance of TM4SF5 as a potential biomarker was evaluated using human HCC formalin-fixed paraffin-embedded tissue samples. The results of the present study demonstrated that low TM4SF5 expression was associated with tumor malignant progression and may be a good prognostic biomarker for OS in HCC.
Collapse
Affiliation(s)
- Baojin Xu
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Wu Lv
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Xiaoyan Li
- Department of Pathology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Lina Zhang
- Department of Pathology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Jie Lin
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| |
Collapse
|
15082
|
Zhou J, Zhu J, Jiang G, Feng J, Wang Q. Downregulation of microRNA-4324 promotes the EMT of esophageal squamous-cell carcinoma cells via upregulating FAK. Onco Targets Ther 2019; 12:4595-4604. [PMID: 31354293 PMCID: PMC6572774 DOI: 10.2147/ott.s198333] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 03/29/2019] [Indexed: 12/19/2022] Open
Abstract
Background: Esophageal squamous-cell carcinoma (ESCC) metastasis is the major cause of death of this severe and common malignancy. Focal adhesion kinase (FAK) is one of the key components of the focal adhesion complex, which is a multi-protein structure that controls cell adhesion, migration and invasion and regulates tumor metastasis. Purpose: To identify the roles and mechanisms of FAK in the regulation of Epithelial-to-mesenchymal transition (EMT) of ESCC cells. Methods: The expression of FAK and miR-4324 in both ESCC tissues and cells were evaluated by qRT-PCR and Immunohistochemistry analysis. Dual luciferase assays were performed for the confirmation of miR-4324's specific binding to 3'UTR of FAK mRNA. Besides, the trans-well assays and wound healing assays were employed to evaluate the effects of FAK /miR-4324 axis on the EMT regulation of ESCC cells. Furthermore, the relationship between miR-4374/FAK expression and clinical pathologic parameters & patient survival were also statistically analyzed. Results: In this study, we identified the upregulation of FAK and downregulation of miR-4324 in both ESCC cells and tissues. Overexpression of miR-4324 mimic, which significantly decreased cellular FAK levels, can impair the invasion potential and migration ability of ESCC cells. Besides, co-transfection of FAK can attenuate the function of miR-4324 mimic. Further experimental results demonstrated that miR-4324 mimic remarkably downregulated epithelial-to-mesenchymal transition (EMT) phenotype, which can also be effectively prevented by overexpressing FAK in ESCC cells. What's more, low miR-4324 and high FAK tissue levels have significant association with poor cell differentiation, tumor size and invasion depth as well as overall number of metastatic lymph nodes. Patients with high miR-4324 and low FAK levels in tumoral tissues lived longer than their counterparts, respectively. Conclusions: In conclusion, miR-4324/FAK axis could be a promising therapeutic target and potential prognostic biomarker for ESCC, which deserves further investigation in the clinic.
Collapse
Affiliation(s)
- Jian Zhou
- Department of Thoracic and Cardiovascular Surgery, Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu Province, 214200, People’s Republic of China
| | - Jiangtao Zhu
- Department of Thoracic and Cardiovascular Surgery, Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu Province, 214200, People’s Republic of China
| | - Guojun Jiang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu Province, 214200, People’s Republic of China
| | - Juncheng Feng
- Department of Thoracic and Cardiovascular Surgery, Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu Province, 214200, People’s Republic of China
| | - Qianqian Wang
- Department of Oncology, Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu Province, 214200, People’s Republic of China
| |
Collapse
|
15083
|
Charo LM, Plaxe SC. Recent advances in endometrial cancer: a review of key clinical trials from 2015 to 2019. F1000Res 2019; 8:F1000 Faculty Rev-849. [PMID: 31231511 PMCID: PMC6567288 DOI: 10.12688/f1000research.17408.1] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/31/2019] [Indexed: 01/08/2023] Open
Abstract
In the past few years, we have seen several important advances in understanding of and therapy for endometrial cancer. This review highlights key recent abstracts and publications in endometrial cancer from 2015 to 2019. We focus on clinical trials in surgical staging and the utility of sentinel lymph node mapping, adjuvant treatment for high-risk disease and HER2/neu-positive serous tumors, combination therapy for recurrent disease, molecular biology, and immunotherapy.
Collapse
Affiliation(s)
- Lindsey M. Charo
- Rebecca and John Moores UC San Diego Cancer Center, 3855 Health Sciences Drive #0987, La Jolla, CA, 92093-0987, USA
| | - Steven C. Plaxe
- Rebecca and John Moores UC San Diego Cancer Center, 3855 Health Sciences Drive #0987, La Jolla, CA, 92093-0987, USA
| |
Collapse
|
15084
|
Shi J, Ma H, Wang H, Zhu W, Jiang S, Dou R, Yan B. Overexpression of LINC00261 inhibits non-small cell lung cancer cells progression by interacting with miR-522-3p and suppressing Wnt signaling. J Cell Biochem 2019; 120:18378-18387. [PMID: 31190356 DOI: 10.1002/jcb.29149] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 05/06/2019] [Accepted: 05/10/2019] [Indexed: 12/25/2022]
Abstract
Long noncoding RNA LINC00261 has been experimentally validated to function as a tumor suppressor in several cancers, but its pathological role and functional mechanism in non-small cell lung cancer (NSCLC) are largely unclear. In this study, LINC00261 was delineated in NSCLC to be significantly downregulated in cancer tissues compared with corresponding adjacent normal tissues. Low expression of LINC00261 predicted worse survival for patients with NSCLC. Overexpression of LINC00261 in NSCLC cell lines inhibited cell proliferation and invasion, meanwhile promoted apoptosis. Subcellular fractionation assay showed that LINC00261 existed mainly in the cytoplasm of NSCLC A549 cells and luciferase assay validated its direct interaction with miR-522-3p. Overexpression of miR-522-3p significantly ameliorated suppressive effects of LINC00261 on proliferation and invasion of NSCLC cells. Besides, miR-522-3p was found to be able to directly combine with the 3'-untranslated region of SFRP2, which was generally regarded as a suppressor of Wnt signaling. Further quantitative reverse transcription polymerase chain reaction and Western blot experiments showed that LINC00261 upregulation potentiated the expression of SFRP2 and inhibited Wnt signaling pathway, which could both be reversely modulated by miR-522-3p. Taken together, our study demonstrated that LINC00261 suppressed NSCLC cells progression via sponging miR-522-3p and inhibiting Wnt signaling. These results supported us to better understand the pathogenic mechanism of NSCLC and revealed a potential molecular target for this fatal disease.
Collapse
Affiliation(s)
- Jingli Shi
- Department of Blood Transfusion, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, China
| | - Huimin Ma
- Department of Blood Transfusion, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, China
| | - Huaixi Wang
- Department of Spine and Spinal Cord Surgery, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, China
| | - Weiyan Zhu
- Department of Blood Transfusion, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, China
| | - Shuting Jiang
- Department of Blood Transfusion, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, China
| | - Rui Dou
- Department of Blood Transfusion, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, China
| | - Beizhan Yan
- Department of Blood Transfusion, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
15085
|
Joe BN, Hayward JH. More Lives Risked with Risk-based versus Age-based Breast Cancer Screening. Radiology 2019; 292:329-330. [PMID: 31184982 DOI: 10.1148/radiol.2019191040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Bonnie N Joe
- Form the Department of Radiology and Biomedical Imaging, University of California San Francisco, 1600 Divisadero St, Room C250, Box 1667, San Francisco, CA 94115
| | - Jessica H Hayward
- Form the Department of Radiology and Biomedical Imaging, University of California San Francisco, 1600 Divisadero St, Room C250, Box 1667, San Francisco, CA 94115
| |
Collapse
|
15086
|
Recio-Boiles A, Nallagangula A, Veeravelli S, Vondrak J, Saboda K, Roe D, Elquza E, McBride A, Babiker HM. Neutrophil-to-lymphocyte and platelet-to-lymphocyte ratios inversely correlate to clinical and pathologic stage in patients with resectable pancreatic ductal adenocarcinoma. ACTA ACUST UNITED AC 2019; 2. [PMID: 31360919 DOI: 10.21037/apc.2019.06.01] [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: 12/17/2022]
Abstract
Background Post-surgical pathology (SP) staging correlates with long-term survival. Neutrophil-lymphocyte ratio (NLR) and platelet-lymphocyte ratio (PLR) have been shown to predict prognosis and extent of tumor in patients with metastatic pancreatic ductal adenocarcinoma (PDAC). This study aimed to correlate NLR and PLR to radiological clinical staging (CS), carbohydrate antigen (CA) 19-9 tumor marker and SP staging in patients with resectable-PDAC (R-PDAC); and to investigate NLR and PLR as potential markers to guide neoadjuvant therapy. Methods Data were collected retrospectively from R-PDAC patients who received upfront surgery from November 2011 to December 2016. NLR and PLR values on the day of diagnosis and surgery were collected. SP, tumor size, location, resected margins (RM), lymphovascular/perineural invasion (LVI/PNI), lymph node involvement, and AJCC/TNM 8th Edition staging were obtained. Associations were assessed using linear, ordinal logistic, and poison regressions or Kruskal Willis Rank Sum Test per the nature of outcome variables, with statistical significance at p-value <0.05. Results Fifty-five patients were identified with resectable stage I (61%) and II (38%). They had a mean age of 66 years (48-87 years) and were 47.2% male, 83.6% white, 90.9% non-Hispanic and 89% with ECOG 0-1. NLR/PLR at diagnosis for R0, R1 and R2 were 6.7/241, 4.8/224, and 2.9/147 (P=0.01/0.002), respectively. NLR/PLR for N0 and N1 were 5.1/212 and 2.7/138.3 (P=0.03/0.009) at diagnosis. No other significant association was detected. Conclusions These findings suggest that NLR/PLR inversely correlates with RM and lymph node status in patients with R-PDAC, but require prospective evaluation in clinically defined scenarios.
Collapse
Affiliation(s)
| | - Aparna Nallagangula
- Creighton University School of Medicine/St. Joseph's Medical Center (Phoenix), Phoenix, AZ, USA
| | | | - Jessica Vondrak
- Department of Internal Medicine, University of Arizona, Tucson, AZ, USA
| | | | - Denise Roe
- University of Arizona College of Public Health, Tucson, AZ, USA
| | - Emad Elquza
- University of Arizona Cancer Center, Tucson, AZ, USA
| | - Ali McBride
- University of Arizona College of Pharmacy, Tucson, AZ, USA
| | | |
Collapse
|
15087
|
Zhang C, Su P, Chen W, Li Q, Dai R, Cheng Y, Yang J. Genetic polymorphisms in IL-7 and IL-7R are correlated with lung cancer risk in the Chinese Han population. Cancer Manag Res 2019; 11:5393-5401. [PMID: 31354347 PMCID: PMC6572729 DOI: 10.2147/cmar.s202839] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/30/2019] [Indexed: 12/14/2022] Open
Abstract
Purpose IL-7/IL-7R axis participates in the initiation and progression of lung cancer (LC). This study aimed to explore the potential influence of IL-7/IL-7R polymorphisms on LC risk. Patients and methods In total, 1,010 participants (507 LC patients and 503 healthy controls) were enrolled. Five single-nucleotide polymorphisms (SNPs) in IL-7R and one SNP in IL-7 were genotyped in included samples with Agena MassARRAY system. OR and 95% CIs were computed by logistic regression analysis after adjusting for age and gender. Stratified analyses with demographic and clinical characteristics were also performed. Finally, linkage disequilibrium (LD) analysis was conducted with the PLINK version 1.07 software . Results IL-7R rs10053847 variant was related to a decreased LC risk under the allele gene (OR =0.78, P=0.043) and additive model (OR =0.77, P=0.042). The results of stratified analysis indicated that this SNP was associated with a lower LC risk among nonsmokers (AA/GG: OR =0.09, P=0.033; AA/AG+GG: OR =0.10 P=0.037) or nondrinkers (AA/GG: OR =0.07, P=0.047; AA/AG+GG: OR =0.18 P=0.049). Moreover, carriers of IL-7R rs10213865-C allele had an increased lung adenocarcinoma risk (CA/AA: OR =1.60, P=0.011; CC+CA/AA: OR =1.62, P=0.007; CA/CA/AA: OR =1.50, P=0.007). Additionally, AGAA haplotype (rs10213865, rs969129, rs118137916 and rs10053847) increased LC risk (OR =1.30, P=0.041). Conclusion IL-7R rs10053847 was correlated with a decreased LC risk, while IL-7R rs10213865 was correlated with an elevated lung adenocarcinoma risk, implying these two SNPs might play essential roles in LC risk evaluation.
Collapse
Affiliation(s)
- Chan Zhang
- Department of Blood Transfusion, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, People's Republic of China
| | - Pincan Su
- Laboratory of Blood Transfusion, Yunnan Kunming Blood Center, Kunming, Yunnan 650106, People's Republic of China
| | - Wanlu Chen
- Department of Blood Transfusion, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, People's Republic of China
| | - Qi Li
- Department of Blood Transfusion, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, People's Republic of China
| | - Run Dai
- Department of Blood Transfusion, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, People's Republic of China
| | - YuJing Cheng
- Department of Blood Transfusion, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, People's Republic of China
| | - Jiangcun Yang
- Department of Transfusion Medicine, Shanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, People's Republic of China
| |
Collapse
|
15088
|
He Y, Deng F, Zhao S, Zhong S, Zhao J, Wang D, Chen X, Zhang J, Hou J, Zhang W, Ding L, Tang J, Zhou Z. Analysis of miRNA-mRNA network reveals miR-140-5p as a suppressor of breast cancer glycolysis via targeting GLUT1. Epigenomics 2019; 11:1021-1036. [PMID: 31184216 DOI: 10.2217/epi-2019-0072] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aim: Aerobic glycolysis is characteristic of breast cancer. Comprehensive expression profiles of key proteins, their prognosis and detailed relationships between miRNAs and mRNAs remain unclear. Materials & methods: Oncomine database, Kaplan-Meier overall survival and miRNA-mRNA network analysis were performed. A key miRNA was identified and explored in vitro and in vivo. Results & conclusion: Eleven key glycolytic proteins were found with higher expression and poor prognosis: GLUT1, SLC2A5, HK1, PFKP, ALDOA, TPI1, GAPDH, PGK1, ENO1, GOT1 and GOT2. Seven miRNAs were predicted targeting 11 key glycolytic proteins: miR-140-5p, miR-3064-5p, miR-152-3p, miR-449b-5p, miR-449a, miR-194-5p and miR-34a-5p. Among them, miR-140-5p was found to be downregulated in breast cancer and directly targeted GLUT1, resulting in an antiglycolytic and antiproliferative effect.
Collapse
Affiliation(s)
- Yunjie He
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Fei Deng
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Shujie Zhao
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210019, PR China
| | - Shanliang Zhong
- Center of Clinical Laboratory, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, PR China
| | - Jianhua Zhao
- Center of Clinical Laboratory, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, PR China
| | - Dandan Wang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Xiu Chen
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Jian Zhang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Junchen Hou
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Wei Zhang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Li Ding
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Jinhai Tang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Zuomin Zhou
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 210029, PR China
| |
Collapse
|
15089
|
Pan H, Wang Y, Na K, Wang Y, Wang L, Li Z, Guo C, Guo D, Wang X. Autophagic flux disruption contributes to Ganoderma lucidum polysaccharide-induced apoptosis in human colorectal cancer cells via MAPK/ERK activation. Cell Death Dis 2019; 10:456. [PMID: 31186406 PMCID: PMC6560101 DOI: 10.1038/s41419-019-1653-7] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/30/2019] [Accepted: 05/13/2019] [Indexed: 01/02/2023]
Abstract
Targeting autophagy may serve as a promising strategy for cancer therapy. Ganoderma lucidum polysaccharide (GLP) has been shown to exert promising anti-cancer effects. However, the underlying mechanisms remain elusive. Whether GLP regulates autophagy in cancer has never been reported. In this study, GLP induced the initiation of autophagy in colorectal cancer (CRC) HT-29 and HCT116 cells, as evidenced by enhanced level of LC3-II protein, GFP-LC3 puncta, and increased formation of double membrane vacuoles. However, GLP treatment caused marked increase of p62 expression. Addition of late stage autophagy inhibitor, chloroquine (CQ), further enhanced LC3-II and p62 level, as well as increased autophagosome accumulation, suggesting a blockage of autophagic flux by GLP in CRC cells. We then found GLP blocked autophagosome and lysosome fusion as determined by mRFP-GFP-LC3 colocalization analysis. Mechanistic study revealed that GLP-induced disruption of autophagosome-lysosome fusion is due to reduced lysosome acidification and lysosomal cathepsin activities. Cell viability and flow cytometry assays revealed that GLP-induced autophagosome accumulation is responsible for GLP-induced apoptosis in CRC cells. In line with this, inhibition of autophagy initiation by 3-methyladenine (3-MA), an early stage autophagy inhibitor, attenuated GLP-induced apoptosis. In contrast, suppression of autophagy at late stage by CQ enhanced the anti-cancer effect of GLP. Furthermore, we demonstrated that GLP-induced autophagosome accumulation and apoptosis is mediated via MAPK/ERK activation. Finally, GLP inhibited tumor growth and also inhibited autophagic flux in vivo. These results unveil new molecular mechanism underlying anti-cancer effects of GLP, suggesting that GLP is a potent autophagy inhibitor and might be useful in anticancer therapy.
Collapse
Affiliation(s)
- Haitao Pan
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Yujie Wang
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Kun Na
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Ying Wang
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Lu Wang
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Zhenhao Li
- Zhejiang Shouxiangu Institute of Rare Medicine Plant, 12, Huanglong 3rd Road, 321200, Wuyi, Zhejiang, China
| | - Chengjie Guo
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Dandan Guo
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Xingya Wang
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China.
| |
Collapse
|
15090
|
Wang H, Kaur G, Sankin AI, Chen F, Guan F, Zang X. Immune checkpoint blockade and CAR-T cell therapy in hematologic malignancies. J Hematol Oncol 2019; 12:59. [PMID: 31186046 PMCID: PMC6558778 DOI: 10.1186/s13045-019-0746-1] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/27/2019] [Indexed: 12/27/2022] Open
Abstract
Harnessing the power of the immune system to recognize and eliminate cancer cells is a longtime exploration. In the past decade, monoclonal antibody (mAb)-based immune checkpoint blockade (ICB) and chimeric antigen receptor T (CAR-T) cell therapy have proven to be safe and effective in hematologic malignancies. Despite the unprecedented success of ICB and CAR-T therapy, only a subset of patients can benefit partially due to immune dysfunction and lack of appropriate targets. Here, we review the preclinical and clinical advances of CTLA-4 and PD-L1/PD-1-based ICB and CD19-specific CAR-T cell therapy in hematologic malignancies. We also discuss the basic research and ongoing clinical trials on emerging immune checkpoints (Galectin-9/Tim-3, CD70/CD27, LAG-3, and LILRBs) and on new targets for CAR-T cell therapy (CD22, CD33, CD123, BCMA, CD38, and CD138) for the treatment of hematologic malignancies.
Collapse
Affiliation(s)
- Hao Wang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Gurbakhash Kaur
- Department of Medical Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Alexander I Sankin
- Department of Urology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Fuxiang Chen
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Department of Medical Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Department of Urology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
15091
|
Yang X, Zhang Q, Zhang M, Su W, Wang Z, Li Y, Zhang J, Beer DG, Yang S, Chen G. Serum microRNA Signature Is Capable of Early Diagnosis for Non-Small Cell Lung Cancer. Int J Biol Sci 2019; 15:1712-1722. [PMID: 31360113 PMCID: PMC6643220 DOI: 10.7150/ijbs.33986] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/23/2019] [Indexed: 01/06/2023] Open
Abstract
Despite decades of efforts, non-small-cell lung cancer (NSCLC) remains the leading cause of cancer mortality globally primarily due to the challenge in early detection of the cancer. Being an important player in cancer development, the dysregulated miRNAs have been shown promising values as non-invasive diagnostic and prognostic biomarkers for NSCLC. The aim of our study is to access the efficacy and reliability of a potential circulating miRNA panel in early diagnosis of NSCLC. We first selected eight candidate miRNAs, miR-146b, miR-205, miR-29c, miR-31, miR-30b, miR-337, miR-411, and miR-708, which have been shown frequently aberrant in primary NSCLC patients based on our previous studies and other reports. The serum level of each of these miRNAs was evaluated by quantitative real-time PCR (qRT-PCR) in training and testing sets. We found that 5 out of 8 miRNAs (miR-146b, miR-205, miR-29c, miR-30b, and miR-337) were significantly up-regulated in NSCLCs patients compared to healthy or cancer-free controls in both training and testing sets. Based on the logistic regression model, a 4-miRNAs set (miR-146b, miR-205, miR-29c and miR-30b) was picked out of the 5 miRNAs owing to its excellent diagnostic power for NSCLC patients in the training set (AUC=0.99, accuracy=95.00%), the testing set (AUC=0.93, accuracy=89.69%), and the training-testing combined set ( AUC=0.96, accuracy=92.00%). When pathological subtypes of NSCLC are compared, this 4-miRNA panel carried a relatively higher prediction power and higher sensitivity for adenocarcinoma (AC) (AUC=0.98, sensitivity=99.10%) than for squamous cell carcinoma (SCC) (AUC=0.93, sensitivity=90.32%). Additionally, this panel demonstrated a comparable diagnostic capacity for stage I (AUC=0.96) and stage II-III (AUC=0.95) of NSCLC, suggesting its role in reflecting the tumor load. Importantly, the high levels of miR-146b and miR-29c in serum were significantly associated with poor 5-year overall survival (OS) (both p=0.04). Further survival analysis showed that high level of miR-146b in serum is specifically correlated with poor survival rate in SCC patients (p=0.0035) but not in AC patients (p=0.83), consistent with our previous finding that the high tissue expression of miR-146b in lung cancer specimen is indicative of a poor prognosis for SCC patients. Altogether, our study demonstrated that the 4-miRNA panel is a novel, sensitive and non-invasive serum marker for the early diagnosis of NSCLC.
Collapse
Affiliation(s)
- Xia Yang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi, China
| | - Qiuhong Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi, China
| | - Ming Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi, China
| | - Wenmei Su
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhuwen Wang
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Yali Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi, China
| | - Jie Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi, China
| | - David G Beer
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Shuanying Yang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi, China
| | - Guoan Chen
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
15092
|
Huang H, Huang Q, Tang T, Zhou X, Gu L, Lu X, Liu F. Differentially Expressed Gene Screening, Biological Function Enrichment, and Correlation with Prognosis in Non-Small Cell Lung Cancer. Med Sci Monit 2019; 25:4333-4341. [PMID: 31181055 PMCID: PMC6582684 DOI: 10.12659/msm.916962] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background The aim of this study was to explore the differently expressed genes and pathways in non-small cell lung cancer (NSCLC) and their correlation with the prognosis. Material/Methods Gene expression data series of GSE19804, GSE101929, and GSE33532 were downloaded from the Gene Expression Ominibus (GEO) database. The overlaping differently expressed genes (DEGs) were identified form the above 3 data series. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEEG) were used to analyze the biological functions and signal pathways of DEGs. The protein–protein interaction (PPI) was analyzed thorough Search Tool for the Retrieval of Interacting Gens (STRING). The relationship between the expression of hub genes and the prognosis of patients was analyzed by Kaplan-Meier Plotter online software. Results Twenty-nine DEGs were identified, with 22 upregulated genes and 7 downregulated genes. The enriched biological processes were mainly related to diet-induced thermogenesis and actin filament binding. The KEGG pathways were enriched in calcium signaling, regulation of lipolysis in adipocytes, and PPAR signaling. Two downregulated genes (MMP1 and SPP1) were identified as hub genes by Cytohubba. Twenty-two dysregulated genes were correlated with patient prognosis. Conclusions Differentially expressed genes are common in NSCLC patients and can be used as biomarkers for patient prognosis.
Collapse
Affiliation(s)
- He Huang
- Department of Respiratory, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Qingdong Huang
- Department of Respiratory, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Tingyu Tang
- Department of Respiratory, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Xiaoxi Zhou
- Department of Respiratory, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Liang Gu
- Department of Respiratory, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Xiaoling Lu
- Department of Respiratory, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Fang Liu
- Department of Respiratory, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
15093
|
Fararjeh AS, Liu YN. ZBTB46, SPDEF, and ETV6: Novel Potential Biomarkers and Therapeutic Targets in Castration-Resistant Prostate Cancer. Int J Mol Sci 2019; 20:E2802. [PMID: 31181727 PMCID: PMC6600524 DOI: 10.3390/ijms20112802] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/25/2019] [Accepted: 06/04/2019] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer (PCa) is the second most common killer among men in Western countries. Targeting androgen receptor (AR) signaling by androgen deprivation therapy (ADT) is the current therapeutic regime for patients newly diagnosed with metastatic PCa. However, most patients relapse and become resistant to ADT, leading to metastatic castration-resistant PCa (CRPC) and eventually death. Several proposed mechanisms have been proposed for CRPC; however, the exact mechanism through which CRPC develops is still unclear. One possible pathway is that the AR remains active in CRPC cases. Therefore, understanding AR signaling networks as primary PCa changes into metastatic CRPC is key to developing future biomarkers and therapeutic strategies for PCa and CRPC. In the current review, we focused on three novel biomarkers (ZBTB46, SPDEF, and ETV6) that were demonstrated to play critical roles in CRPC progression, epidermal growth factor receptor tyrosine kinase inhibitor (EGFR TKI) drug resistance, and the epithelial-to-mesenchymal transition (EMT) for patients treated with ADT or AR inhibition. In addition, we summarize how these potential biomarkers can be used in the clinic for diagnosis and as therapeutic targets of PCa.
Collapse
Affiliation(s)
- AbdulFattah Salah Fararjeh
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yen-Nien Liu
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
- Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
15094
|
Eberle-Singh JA, Sagalovskiy I, Maurer HC, Sastra SA, Palermo CF, Decker AR, Kim MJ, Sheedy J, Mollin A, Cao L, Hu J, Branstrom A, Weetall M, Olive KP. Effective Delivery of a Microtubule Polymerization Inhibitor Synergizes with Standard Regimens in Models of Pancreatic Ductal Adenocarcinoma. Clin Cancer Res 2019; 25:5548-5560. [PMID: 31175095 DOI: 10.1158/1078-0432.ccr-18-3281] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 03/26/2019] [Accepted: 06/03/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDA) is a deadly cancer that is broadly chemoresistant, due in part to biophysical properties of tumor stroma, which serves as a barrier to drug delivery for most classical chemotherapeutic drugs. The goal of this work is to evaluate the preclinical efficacy and mechanisms of PTC596, a novel agent with potent anticancer properties in vitro and desirable pharmacologic properties in vivo.Experimental Design: We assessed the pharmacology, mechanism, and preclinical efficacy of PTC596 in combination with standards of care, using multiple preclinical models of PDA. RESULTS We found that PTC596 has pharmacologic properties that overcome the barrier to drug delivery in PDA, including a long circulating half-life, lack of P-glycoprotein substrate activity, and high systemic tolerability. We also found that PTC596 combined synergistically with standard clinical regimens to improve efficacy in multiple model systems, including the chemoresistant genetically engineered "KPC" model of PDA. Through mechanistic studies, we learned that PTC596 functions as a direct microtubule polymerization inhibitor, yet a prior clinical trial found that it lacks peripheral neurotoxicity, in contrast to other such agents. Strikingly, we found that PTC596 synergized with the standard clinical backbone regimen gemcitabine/nab-paclitaxel, yielding potent, durable regressions in a PDX model. Moreover, similar efficacy was achieved in combination with nab-paclitaxel alone, highlighting a specific synergistic interaction between two different microtubule-targeted agents in the setting of pancreatic ductal adenocarcinoma. CONCLUSIONS These data demonstrate clear rationale for the development of PTC596 in combination with standard-of-care chemotherapy for PDA.
Collapse
Affiliation(s)
- Jaime A Eberle-Singh
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, New York.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Irina Sagalovskiy
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, New York.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - H Carlo Maurer
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, New York.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Stephen A Sastra
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, New York.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Carmine F Palermo
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, New York.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Amanda R Decker
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, New York.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | | | | | - Anna Mollin
- PTC Therapeutics, South Plainfield, New Jersey
| | | | - Jianhua Hu
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York.,Department of Biostatistics, Columbia University Medical Center, New York, New York
| | | | | | - Kenneth P Olive
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, New York. .,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| |
Collapse
|
15095
|
Li L, Li Y, Yin Z, Zhu J, Yan D, Lou H. Increased frequency of regulatory T cells in the peripheral blood of patients with endometrioid adenocarcinoma. Oncol Lett 2019; 18:1424-1430. [PMID: 31423207 DOI: 10.3892/ol.2019.10452] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 04/05/2019] [Indexed: 12/23/2022] Open
Abstract
The aim of the present study was to investigate the frequency of cluster of differentiation (CD)4+CD25+CD127- regulatory T cells (Tregs) in the peripheral blood mononuclear cells (PBMCs) of patients with endometrioid adenocarcinoma (EA). A total of 82 female patients with EA were recruited. The PBMCs were flow cytometrically analyzed to determine the percentage of CD4+CD25+CD127- Tregs within the CD4+ T cell population. The associations between the prevalence of Tregs in PBMCs and defined clinical prognostic parameters were evaluated. To study the immunoregulatory capacity of Tregs, the level of specific cytokines were detected by ELISA, and the proliferation of cells was analyzed by incorporation of 3H-thymidine. It was revealed that Treg/CD4+ ratio in the peripheral blood of patients with EA was 4.89±1.42%, significantly higher than Treg/CD4+ ratio in healthy women. No correlation was observed between Treg frequency and stage, grade of differentiation, menopausal status or age. CD4+CD25+CD127- Tregs secreted large amounts of IL-10 but not IFN-γ. The level of IL-10 secreted by Tregs from patients with EA and healthy controls was not significantly different. In addition, there was no significant difference in the suppressive activity of Tregs in patients with EA compared with that of the healthy controls. These findings demonstrate that the increased frequency of immunosuppressive Tregs in patients with EA may be responsible for immune tolerance in endometrial cancer.
Collapse
Affiliation(s)
- Li Li
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China.,Key Laboratory of Radiation Oncology of Zhejiang Province, Hangzhou, Zhejiang 310022, P.R. China
| | - Yinghua Li
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Zhuomin Yin
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Jing Zhu
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Dingding Yan
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Hanmei Lou
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China.,Key Laboratory of Radiation Oncology of Zhejiang Province, Hangzhou, Zhejiang 310022, P.R. China
| |
Collapse
|
15096
|
Khalili M, Daniels L, Gleeson EM, Grandhi N, Thandoni A, Burg F, Holleran L, Morano WF, Bowne WB. Pancreaticoduodenectomy outcomes for locally advanced right colon cancers: A systematic review. Surgery 2019; 166:223-229. [PMID: 31182232 DOI: 10.1016/j.surg.2019.04.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/08/2019] [Accepted: 04/24/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND Pancreaticoduodenectomy (PD) with right hemicolectomy (RH) to treat locally advanced right colon cancer (LARCC) has been rarely reported in the literature. Herein, we characterize clinicopathologic factors and evaluate outcomes of en bloc PD and RH for LARCC. METHODS A systematic review of the literature was conducted on PubMed using MeSH terms ("pancreaticoduodenectomy" or "pancreas/surgery" or "duodenum/surgery" or "colectomy") and ("colonic neoplasms"). Data was extracted from patients who underwent en bloc PD and RH for LARCC. Factors investigated included patient demographics, surgical and pathologic parameters, postoperative complications, disease recurrence, and survival. RESULTS Our search yielded 27 articles (106 patients), including 1 case from our institution. Most patients were male (62.1%), median age 58 years (range 34-83). Surgical procedures performed included en bloc RH with PD (n = 91, 85.8%) and en bloc RH with pylorus-preserving PD (n = 15, 14.2%). Among reported, 95.5% of patients (n = 63), underwent R0 resection. One or more complications were reported in 33 patients (52.4%). Median survival was 168 months. Survival after resection was 75.9% at 2 years and 66.3% at 5 years. Overall survival was greater in patients with no lymph node involvement (IIC versus IIIC, hazard ratio 8.4, P = .003). Five-year survival for patients was 84.9% in patients with stage IIC versus 46.4% in patients with stage IIIC. There were 3 postoperative mortalities. CONCLUSION This data demonstrates that en bloc PD and RH is rarely performed yet can be a potentially safe treatment option in patients with LARCC. Lymph node involvement was the only independent prognostic factor.
Collapse
Affiliation(s)
- Marian Khalili
- Department of Surgery, Hahnemann University Hospital and Drexel University College of Medicine, Philadelphia, PA
| | - Lynsey Daniels
- Department of Surgery, Hahnemann University Hospital and Drexel University College of Medicine, Philadelphia, PA
| | - Elizabeth M Gleeson
- Department of Surgery, Hahnemann University Hospital and Drexel University College of Medicine, Philadelphia, PA
| | - Nikhil Grandhi
- Department of Surgery, Hahnemann University Hospital and Drexel University College of Medicine, Philadelphia, PA
| | - Aditya Thandoni
- Department of Surgery, Hahnemann University Hospital and Drexel University College of Medicine, Philadelphia, PA
| | - Franklin Burg
- Department of Surgery, Hahnemann University Hospital and Drexel University College of Medicine, Philadelphia, PA
| | - Lauren Holleran
- Department of Surgery, Hahnemann University Hospital and Drexel University College of Medicine, Philadelphia, PA
| | - William F Morano
- Department of Surgery, Hahnemann University Hospital and Drexel University College of Medicine, Philadelphia, PA
| | - Wilbur B Bowne
- Department of Surgery, Hahnemann University Hospital and Drexel University College of Medicine, Philadelphia, PA.
| |
Collapse
|
15097
|
Uthoff J, Stephens MJ, Newell JD, Hoffman EA, Larson J, Koehn N, De Stefano FA, Lusk CM, Wenzlaff AS, Watza D, Neslund-Dudas C, Carr LL, Lynch DA, Schwartz AG, Sieren JC. Machine learning approach for distinguishing malignant and benign lung nodules utilizing standardized perinodular parenchymal features from CT. Med Phys 2019; 46:3207-3216. [PMID: 31087332 DOI: 10.1002/mp.13592] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 04/25/2019] [Accepted: 05/07/2019] [Indexed: 11/09/2022] Open
Abstract
PURPOSE Computed tomography (CT) is an effective method for detecting and characterizing lung nodules in vivo. With the growing use of chest CT, the detection frequency of lung nodules is increasing. Noninvasive methods to distinguish malignant from benign nodules have the potential to decrease the clinical burden, risk, and cost involved in follow-up procedures on the large number of false-positive lesions detected. This study examined the benefit of including perinodular parenchymal features in machine learning (ML) tools for pulmonary nodule assessment. METHODS Lung nodule cases with pathology confirmed diagnosis (74 malignant, 289 benign) were used to extract quantitative imaging characteristics from computed tomography scans of the nodule and perinodular parenchyma tissue. A ML tool development pipeline was employed using k-medoids clustering and information theory to determine efficient predictor sets for different amounts of parenchyma inclusion and build an artificial neural network classifier. The resulting ML tool was validated using an independent cohort (50 malignant, 50 benign). RESULTS The inclusion of parenchymal imaging features improved the performance of the ML tool over exclusively nodular features (P < 0.01). The best performing ML tool included features derived from nodule diameter-based surrounding parenchyma tissue quartile bands. We demonstrate similar high-performance values on the independent validation cohort (AUC-ROC = 0.965). A comparison using the independent validation cohort with the Fleischner pulmonary nodule follow-up guidelines demonstrated a theoretical reduction in recommended follow-up imaging and procedures. CONCLUSIONS Radiomic features extracted from the parenchyma surrounding lung nodules contain valid signals with spatial relevance for the task of lung cancer risk classification. Through standardization of feature extraction regions from the parenchyma, ML tool validation performance of 100% sensitivity and 96% specificity was achieved.
Collapse
Affiliation(s)
- Johanna Uthoff
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52240, USA.,Department of Radiology, University of Iowa, Iowa City, IA, 52242, USA
| | - Matthew J Stephens
- Department of Radiology, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - John D Newell
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52240, USA.,Department of Radiology, University of Iowa, Iowa City, IA, 52242, USA
| | - Eric A Hoffman
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52240, USA.,Department of Radiology, University of Iowa, Iowa City, IA, 52242, USA
| | - Jared Larson
- Department of Radiology, University of Iowa, Iowa City, IA, 52242, USA
| | - Nicholas Koehn
- Department of Radiology, University of Iowa, Iowa City, IA, 52242, USA
| | | | - Chrissy M Lusk
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Angela S Wenzlaff
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Donovan Watza
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | | | - Laurie L Carr
- Department of Medicine, National Jewish Health, Denver, CO, 80206, USA
| | - David A Lynch
- Department of Radiology, National Jewish Health, Denver, CO, 80206, USA
| | - Ann G Schwartz
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Jessica C Sieren
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52240, USA.,Department of Radiology, University of Iowa, Iowa City, IA, 52242, USA
| |
Collapse
|
15098
|
Zhang J, Zhang Y, Ma Z. In silico Prediction of Human Secretory Proteins in Plasma Based on Discrete Firefly Optimization and Application to Cancer Biomarkers Identification. Front Genet 2019; 10:542. [PMID: 31244885 PMCID: PMC6563772 DOI: 10.3389/fgene.2019.00542] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 05/21/2019] [Indexed: 12/20/2022] Open
Abstract
The early control and prevention of cancer contributes effectively interventions and cancer therapies. Secretory protein, one of the richest biomarkers, is proved important as molecular signposts of the physiological state of a cell. In this work, we aim to propose a proteomic high-throughput technology platform to facilitate detection of early cancer by means of biomarkers that secreted into the bloodstream. We compile a new benchmark dataset of human secretory proteins in plasma. A series of sequence-derived features, which have been proved involved in the structure and function of the secretory proteins, are collected to mathematically encode these proteins. Considering the influence of potential irrelevant or redundant features, we introduce discrete firefly optimization algorithm to perform feature selection. We evaluate and compare the proposed method SCRIP (Secretory proteins in plasma) with state-of-the-art approaches on benchmark datasets and independent testing datasets. SCRIP achieves the average AUC values of 0.876 and 0.844 in five-fold the cross-validation and independent test, respectively. Besides that, we also test SCRIP on proteins in four types of cancer tissues and successfully detect 66∼77% potential cancer biomarkers.
Collapse
Affiliation(s)
- Jian Zhang
- School of Computer and Information Technology, Xinyang Normal University, Xinyang, China
- Henan Key Laboratory of Education Big Data Analysis and Application, Xinyang, China
| | - Yu Zhang
- Information Engineering College, Huanghuai University, Zhumadian, China
- Henan Key Laboratory of Smart Lighting, Zhumadian, China
| | - Zhiqiang Ma
- Department of Computer Science, College of Humanities & Sciences of Northeast Normal University, Changchun, China
| |
Collapse
|
15099
|
Zhong J, Ren X, Chen Z, Zhang H, Zhou L, Yuan J, Li P, Chen X, Liu W, Wu D, Yang X, Liu J. miR-21-5p promotes lung adenocarcinoma progression partially through targeting SET/TAF-Iα. Life Sci 2019; 231:116539. [PMID: 31176779 DOI: 10.1016/j.lfs.2019.06.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/29/2019] [Accepted: 06/05/2019] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Although SET(I2PP2A) and miRNAs are reported to play a pivotal role in lung cancer, the underlying mechanisms have remained obscure. To address this issue, we investigated how miRNAs and SET participate in the progression of lung cancer. METHODS miRNAs that target SET were predicted from multiple miRNA databases. Three human NSCLC cell lines and two normal lung cell lines were used to evaluate aberrant miRNA and SET expressions. A dual luciferase reporter assay system was employed to verify the interaction between miRNA and SET. Stable miRNA knockdown and SET overexpression in A549 cells were achieved through lentivirus transfection; the corresponding influences on lung cancer progression were also examined. RESULTS In this study, A549 was the sole cell line to lack SET/TAF-Iα expression, which was inversely correlated with the up-regulation of miR-21-5p. SET was subsequently revealed as the direct target site of miR-21-5p in A549 cells. The stable miR-21-5p knockdown and SET/TAF-Iα overexpression were shown to markedly enhance the expression of SET/TAF-Iα and to inhibit the migration, invasion, proliferation as well as the in vivo tumorigenicity of A549 cells. CONCLUSION We suggest that SET/TAF-Iα might be a tumor suppressing factor regulated by miR-21-5p in lung adenocarcinoma. This might provide a target for lung adenocarcinoma therapy.
Collapse
Affiliation(s)
- Jiacheng Zhong
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Xiaohu Ren
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Zhihong Chen
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Hang Zhang
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Li Zhou
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Jianhui Yuan
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Ping Li
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Xiao Chen
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Wei Liu
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Desheng Wu
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Xifei Yang
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Jianjun Liu
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China.
| |
Collapse
|
15100
|
Lu YT, Delijani K, Mecum A, Goldkorn A. Current status of liquid biopsies for the detection and management of prostate cancer. Cancer Manag Res 2019; 11:5271-5291. [PMID: 31239778 PMCID: PMC6559244 DOI: 10.2147/cmar.s170380] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/18/2019] [Indexed: 12/14/2022] Open
Abstract
In recent years, new therapeutic options have become available for prostate cancer (PC) patients, generating an urgent need for better biomarkers to guide the choice of therapy and monitor treatment response. Liquid biopsies, including circulating tumor cells (CTCs), circulating nucleic acids, and exosomes, have been developed as minimally invasive assays allowing oncologists to monitor PC patients with real-time cellular or molecular information. While CTC counts remain the most extensively validated prognostic biomarker to monitor treatment response, recent advances demonstrate that CTC morphology and androgen receptor characterization can provide additional information to guide the choice of treatment. Characterization of cell-free DNA (cfDNA) is another rapidly emerging field with novel technologies capable of monitoring the evolution of treatment relevant alterations such as those in DNA damage repair genes for poly (ADP-ribose) polymerase (PARP) inhibition. In addition, several new liquid biopsy fields are emerging, including the characterization of heterogeneity, CTC RNA sequencing, the culture and xenografting of CTCs, and the characterization of extracellular vesicles (EVs) and circulating microRNAs. This review describes the clinical utilization of liquid biopsies in the management of PC patients and emerging liquid biopsy technologies with the potential to advance personalized cancer therapy.
Collapse
Affiliation(s)
- Yi-Tsung Lu
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Kevin Delijani
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Andrew Mecum
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Amir Goldkorn
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|