15001
|
Wong MCS, Ding H, Wang J, Chan PSF, Huang J. Prevalence and risk factors of colorectal cancer in Asia. Intest Res 2019; 17:317-329. [PMID: 31085968 PMCID: PMC6667372 DOI: 10.5217/ir.2019.00021] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 02/06/2023] Open
Abstract
Globally, colorectal cancer (CRC) is a substantial public health burden, and it is increasingly affecting populations in Asian countries. The overall prevalence of CRC is reported to be low in Asia when compared with that in Western nations, yet it had the highest number of prevalent cases. This review described the prevalence of CRC in Asia according to the International Agency for Research on Cancer from World Health Organization (WHO) database and summarized its major risk factors. Non-modifiable factors include genetic factors, ethnicity, age, gender, family history and body height; smoking, alcohol drinking, weight, Westernized diet, physical inactivity, chronic diseases and microbiota were involved in environmental factors. These risk factors were separately discussed in this review according to published literature from Asian countries. CRC screening has been playing an important role in reducing its disease burden. Some recommendations on its screening practices have been formulated in guidelines for Asia Pacific countries.
Collapse
Affiliation(s)
- Martin CS Wong
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Hanyue Ding
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jingxuan Wang
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Paul SF Chan
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Junjie Huang
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
15002
|
Chen EY, Raghunathan V, Prasad V. An Overview of Cancer Drugs Approved by the US Food and Drug Administration Based on the Surrogate End Point of Response Rate. JAMA Intern Med 2019; 179:915-921. [PMID: 31135822 PMCID: PMC6547222 DOI: 10.1001/jamainternmed.2019.0583] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
IMPORTANCE Approximately one-third of cancer drugs are approved based on response rate (RR)-the percentage of patients whose tumors shrink beyond an arbitrary threshold-typically assessed in a single-arm study. OBJECTIVE To characterize RR end points used by the US Food and Drug Administration (FDA) for cancer drug approval. DESIGN, SETTING, AND PARTICIPANTS A retrospective review of FDA-approved drug indications in oncology from 2006 to 2018. EXPOSURES Data related to cancer type, line of therapy (first-line, second-line, or third-or-later-line treatment for advanced/metastatic disease), type of FDA approval pathway, trial design, sample size, and level of innovation were extracted. MAIN OUTCOMES AND MEASURES The primary outcome was the RR used as the basis for FDA approval. The secondary outcome was rate of complete response. RESULTS Eighty-five indications for 59 cancer drugs were identified, 32 (38%) received regular approval, and 53 (62%) were granted accelerated approval. Twenty-nine (55%) accelerated approvals were later converted to regular approval. Of these, 6 (21%) approvals showed overall survival benefit, 16 (55%) later established progression-free survival benefit, and 7 (24%) continued to use RR but gained regular approval. The median RR among the 85 indications was 41% (interquartile range [IQR], 27%-58%). Among them, 14 of 85 (16%) had an RR less than 20%, 28 of 85 (33%) had an RR less than 30%, and 40 of 85 (47%) had an RR less than 40%. The median complete RR for 81 participants was 6% (IQR, 2%-22%). The median sample size among studies leading to approval was 117 (IQR, 76-182; range, 18-1052 participants). Drugs with accelerated approval pending confirmatory data had lower RR compared with drugs that have completed most postmarketing efficacy requirements (median, 28%; IQR, 15%-50% vs median, 42%; IQR, 31%-58%; P = .02). CONCLUSIONS AND RELEVANCE Many cancer drugs approved on the basis of response rate offer numerically low or modest response rates. Most premarket studies accrue more than 100 patients. Some of these drugs could potentially be tested in premarket randomized clinical trials measuring directly end points that demonstrate clinical benefit.
Collapse
Affiliation(s)
- Emerson Y Chen
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland
| | - Vikram Raghunathan
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland
| | - Vinay Prasad
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland.,Department of Public Health and Preventive Medicine, Oregon Health & Science University, Portland
| |
Collapse
|
15003
|
Martorana E, Pirola GM, Aisa MC, Scialpi P, Di Blasi A, Saredi G, D'Andrea A, Signore S, Grisanti R, Scialpi M. Prostate MRI and transperineal TRUS/MRI fusion biopsy for prostate cancer detection: clinical practice updates. Turk J Urol 2019; 45:237-244. [PMID: 31291186 DOI: 10.5152/tud.2019.19106] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 06/18/2019] [Indexed: 11/22/2022]
Abstract
This narrative review summarizes the current knowledge about multiparametric and biparametric magnetic resonance imaging of the prostate. This is provided from both a radiological and a urological point of view analyzing the technical aspects of fusion-targeted biopsy using the transperineal approach. We report practical considerations concerning pure cognitive and software-assisted settings, discuss the principal transperineal fusion software now available, and debate the pros and cons of choosing one approach over the other.
Collapse
Affiliation(s)
| | | | - Maria Cristina Aisa
- Division of Obstetrics and Gynecology, Department of Surgical and Biomedical Sciences, University of Perugia, Italy
| | - Pietro Scialpi
- Department of Urology, Portogruaro Hospital, Portogruaro, Italy
| | - Aldo Di Blasi
- Section of Radiology and Diagnostic Imaging, Tivoli Hospital, Lazio, Italy
| | | | | | | | | | - Michele Scialpi
- Division of Diagnostic Imaging, Department of Surgical and Biomedical Sciences, University of Perugia, Italy
| |
Collapse
|
15004
|
Liu B, Huang X, Li Y, Liao W, Li M, Liu Y, He R, Feng D, Zhu R, Kurihara H. JS-K, a nitric oxide donor, induces autophagy as a complementary mechanism inhibiting ovarian cancer. BMC Cancer 2019; 19:645. [PMID: 31262254 PMCID: PMC6604176 DOI: 10.1186/s12885-019-5619-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 04/16/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Ovarian cancer (OC) is the second most frequent gynecological cancer and is associated with a poor prognosis because OC progression is often asymptoma-tic and is detected at a late stage. There remains an urgent need for novel targeted therapies to improve clinical outcomes in ovarian cancer. As a nitric oxide prodrug, JS-K is reported highly cytotoxic to human cancer cells such as acute myeloid leukemia, multiple myeloma and breast cancer. This study is aim to investigate the influence of JS-K on proliferation and apoptosis in ovarian cancer cells and explored possible autophagy-related mechanisms, which will contribute to future ovarian cancer therapy and supply theory support that JS-K holds great promise as a novel therapeutic agent against ovarian cancer. METHODS The cytotoxicity, extracellular ROS/RNS activity and apoptotic effect of JS-K and indicated inhibitors on ovarian cancer cells in vitro were evaluated by MTT assay, extracellular ROS/RNS assay, caspases activities assay and western blot. Further autophagy effect of JS-K and indicated inhibitors were examined by MTT assay, cell transfection, immunofluorescence analysis, transmission electron microscopy (TEM) analysis and western blot on ovarian cancer cells in vitro. In vivo, the BALB/c-nude female mice with SKOV3 ovarian cancer cells xenograft were used to examine the efficacy of JS-K treatment on tumor growth. PCNA and p62 proteins were analyzed by immunohistochemistry. RESULTS In vitro, JS-K inhibited the proliferation of ovarian cancer cells, induced apoptosis and cell nucleus shrinkage, enhanced the enzymatic activity of caspase-3/7/8/9, and significantly increased the production of ROS/RNS in ovarian cancer A2780 and SKOV3 cells, these effects were attenuated by inhibition of NAC. In addition, JS-K induced autophagy-related proteins and autophagosomes changes in ovarian cancer A2780 and SKOV3 cells. In vivo, JS-K inhibited tumor growth, decreased p62 protein expression and increased the expression levels of PCNA in xenograft models which were established using SKOV3 ovarian cancer cells. CONCLUSION Taken together, we demonstrated that ROS/RNS stress-mediated apoptosis and autophagy are mechanisms by which SKOV3 cells undergo cell death after treatment with JS-K in vitro. Moreover, JS-K inhibited SKOV3 tumor growth in vivo. An alternative therapeutic approach for triggering cell death in cancer cells could constitute a useful multimodal therapies for treating ovarian cancer, which is known for its resistance to apoptosis-inducing drugs.
Collapse
Affiliation(s)
- Bin Liu
- College of Pharmacy, Jinan University, Guangzhou, 510632 Guangdong China
- Laboratory of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001 Guangdong China
| | - Xiaojie Huang
- Laboratory of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001 Guangdong China
| | - Yifang Li
- College of Pharmacy, Jinan University, Guangzhou, 510632 Guangdong China
| | - Weiguo Liao
- Laboratory of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001 Guangdong China
| | - Mingyi Li
- Laboratory of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001 Guangdong China
| | - Yi Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016 Liaoning China
| | - Rongrong He
- College of Pharmacy, Jinan University, Guangzhou, 510632 Guangdong China
| | - Du Feng
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital & Institute, Guangzhou, Medical University, Guangzhou, 511436 Guangdong China
| | - Runzhi Zhu
- Laboratory of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001 Guangdong China
- Center for Cell Therapy, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001 Jiangsu China
| | - Hiroshi Kurihara
- College of Pharmacy, Jinan University, Guangzhou, 510632 Guangdong China
| |
Collapse
|
15005
|
Cowey CL, Liu FX, Boyd M, Aguilar KM, Krepler C. Real-world treatment patterns and clinical outcomes among patients with advanced melanoma: A retrospective, community oncology-based cohort study (A STROBE-compliant article). Medicine (Baltimore) 2019; 98:e16328. [PMID: 31305421 PMCID: PMC6641721 DOI: 10.1097/md.0000000000016328] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Recently, the effectiveness of novel immune checkpoint inhibitors and BRAF-directed therapies has been demonstrated in advanced melanoma trial populations. Limited research, however, has evaluated the impact of these therapies in a real-world setting. The aim of this study was to evaluate treatment patterns and clinical outcomes among advanced melanoma patients treated with modern therapies within community oncology clinics. Adult patients with advanced melanoma who initiated treatment within the US Oncology Network between 1/1/14 and 12/31/16 were included. Data were sourced from electronic healthcare records. Patients were followed through 12/31/17. Descriptive analyses were performed to assess patient and treatment characteristics and Kaplan-Meier methods were used for time-to-event outcomes. In total, 484 patients met eligibility criteria (32.0% with brain metastasis, 12.6% with Eastern Cooperative Oncology Group performance status ≥2). In the first-line (1L) setting during the study period, 37.0% received anti-PD1 monotherapies, 26.4% ipilimumab monotherapy, 19.8% BRAF/MEK combination therapy, 6.4% BRAF or MEK monotherapy, 4.1% ipilimumab/nivolumab combination therapy and 6.2% other regimens. Differences in baseline demographic and clinical characteristics were observed across treatment groups. For the overall study population, the median (95% confidence interval) estimates for overall survival, time to next treatment and progression-free survival were 20.7 (16.0, 26.8), 5.8 (5.3, 6.5), and 4.9 (4.2, 5.7) months, respectively. The results of this study provide real-world insight into advanced melanoma treatment trends and clinical outcomes, including high utilization of immunotherapies and BRAF/MEK combination therapy. Future research can explore underlying differences in patient subpopulations and the sequence of therapies across lines of therapy.
Collapse
|
15006
|
Odisho AY, Bridge M, Webb M, Ameli N, Eapen RS, Stauf F, Cowan JE, Washington SL, Herlemann A, Carroll PR, Cooperberg MR. Automating the Capture of Structured Pathology Data for Prostate Cancer Clinical Care and Research. JCO Clin Cancer Inform 2019; 3:1-8. [PMID: 31314550 PMCID: PMC6874052 DOI: 10.1200/cci.18.00084] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2019] [Indexed: 01/19/2023] Open
Abstract
PURPOSE Cancer pathology findings are critical for many aspects of care but are often locked away as unstructured free text. Our objective was to develop a natural language processing (NLP) system to extract prostate pathology details from postoperative pathology reports and a parallel structured data entry process for use by urologists during routine documentation care and compare accuracy when compared with manual abstraction and concordance between NLP and clinician-entered approaches. MATERIALS AND METHODS From February 2016, clinicians used note templates with custom structured data elements (SDEs) during routine clinical care for men with prostate cancer. We also developed an NLP algorithm to parse radical prostatectomy pathology reports and extract structured data. We compared accuracy of clinician-entered SDEs and NLP-parsed data to manual abstraction as a gold standard and compared concordance (Cohen's κ) between approaches assuming no gold standard. RESULTS There were 523 patients with NLP-extracted data, 319 with SDE data, and 555 with manually abstracted data. For Gleason scores, NLP and clinician SDE accuracy was 95.6% and 95.8%, respectively, compared with manual abstraction, with concordance of 0.93 (95% CI, 0.89 to 0.98). For margin status, extracapsular extension, and seminal vesicle invasion, stage, and lymph node status, NLP accuracy was 94.8% to 100%, SDE accuracy was 87.7% to 100%, and concordance between NLP and SDE ranged from 0.92 to 1.0. CONCLUSION We show that a real-world deployment of an NLP algorithm to extract pathology data and structured data entry by clinicians during routine clinical care in a busy clinical practice can generate accurate data when compared with manual abstraction for some, but not all, components of a prostate pathology report.
Collapse
Affiliation(s)
| | - Mark Bridge
- University of California, San Francisco, San Francisco, CA
| | - Mitchell Webb
- University of California, San Francisco Medical Center, San Francisco, CA
| | - Niloufar Ameli
- University of California, San Francisco, San Francisco, CA
| | - Renu S Eapen
- University of California, San Francisco, San Francisco, CA
| | - Frank Stauf
- University of California, San Francisco, San Francisco, CA
| | - Janet E Cowan
- University of California, San Francisco, San Francisco, CA
| | | | | | | | | |
Collapse
|
15007
|
Ma J, Zhang L, Bian HR, Lu ZG, Zhu L, Yang P, Zeng ZC, Xiang ZL. A Noninvasive Prediction Nomogram for Lymph Node Metastasis of Hepatocellular Carcinoma Based on Serum Long Noncoding RNAs. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1710670. [PMID: 31355249 PMCID: PMC6634290 DOI: 10.1155/2019/1710670] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/17/2019] [Accepted: 05/23/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND OBJECTIVES Lymph node metastasis (LNM) is common in hepatocellular carcinoma (HCC). In order to intervene HCC LNM in advance, we developed a prediction nomogram based on serum long noncoding RNA (lncRNA). METHODS Serum samples from 242 HCC patients were gathered and randomly enrolled into the training and validation cohorts. LncRNAs screened out from microarray were quantified with qRT-PCR. Univariate and multivariate analyses were applied for screening independent risk factors. A prediction nomogram was ultimately developed for HCC LNM. The nomogram was estimated by discrimination and calibration tests in the validation cohort. The effects of the candidate lncRNA on the malignant phenotypes of HCC cells were further explored by wound healing assay and colony formation assay. RESULTS ENST00000418803, lnc-ZNF35-4:1, lnc-EPS15L1-2:1, BCLC stage, and vascular invasion were selected as components of the nomogram according to the adjusted multivariate analysis. The nomogram effectively predicted the HCC LNM risk among the cohorts with suitable calibration fittings and displayed high discrimination with C-index of 0.89 and 0.85. Moreover, the abnormally high expression of lnc-EPS15L1-2:1 in HCC cell lines showed significant carcinogenic effects. CONCLUSIONS The noninvasive nomogram may provide more diagnostic basis for treatments of HCC. The biomarkers identified can bring new clues to basic researches.
Collapse
Affiliation(s)
- Jie Ma
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Li Zhang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hai-Rong Bian
- Department of Radiation Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zheng-Guo Lu
- Department of Radiation Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lian Zhu
- Department of Radiation Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ping Yang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhao-Chong Zeng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zuo-Lin Xiang
- Department of Radiation Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
15008
|
The synergistic effect of PFK15 with metformin exerts anti-myeloma activity via PFKFB3. Biochem Biophys Res Commun 2019; 515:332-338. [DOI: 10.1016/j.bbrc.2019.05.136] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 05/21/2019] [Indexed: 02/08/2023]
|
15009
|
LeVan TD, Xiao P, Kumar G, Kupzyk K, Qiu F, Klinkebiel D, Eudy J, Cowan K, Berger AM. Genetic Variants in Circadian Rhythm Genes and Self-Reported Sleep Quality in Women with Breast Cancer. J Circadian Rhythms 2019; 17:6. [PMID: 31303884 PMCID: PMC6611482 DOI: 10.5334/jcr.184] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 06/05/2019] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Women diagnosed with breast cancer (BC) are at increased risk of sleep deficiency. Approximately 30-60% of these women report poor sleep during and following surgery, chemotherapy, radiation therapy, and anti-estrogen therapy. The purpose of this study was to examine the relationship between genetic variation in circadian rhythm genes and self-reported sleep quality in women with BC. METHODS This cross-sectional study recruited women with a first diagnosis of breast cancer at five sites in Nebraska and South Dakota. Sixty women were included in the study. Twenty-six circadian genes were selected for exome sequencing using the Nextera Rapid Capture Expanded Exome kit. 414 variants had a minor allele frequency of ≥5% and were included in the exploratory analysis. The association between Pittsburgh Sleep Quality Index (PSQI) score and genetic variants was determined by two-sample t-test or ANOVA. RESULTS Twenty-five variants were associated with the PSQI score at p < 0.10, of which 19 were significant at p<0.05, although the associations did not reach statistical significance after adjustment for multiple comparisons. Variants associated with PSQI were from genes CSNK1D & E, SKP1, BHLHE40 & 41, NPAS2, ARNTL, MYRIP, KLHL30, TIMELESS, FBXL3, CUL1, PER1&2, RORB. Two genetic variants were synonymous or missense variants in the BHLHE40 and TIMELESS genes, respectively. CONCLUSIONS These exploratory results demonstrate an association of genetic variants in circadian rhythm pathways with self-reported sleep in women with BC. Testing this association is warranted in a larger replication population.
Collapse
Affiliation(s)
| | - Peng Xiao
- University of Nebraska Medical Center, US
| | | | | | - Fang Qiu
- University of Nebraska Medical Center, US
| | | | - James Eudy
- University of Nebraska Medical Center, US
| | | | | |
Collapse
|
15010
|
Xia T, Sun H, Huang H, Bi H, Pu R, Zhang L, Zhang Y, Liu Y, Xu J, Onwuka JU, Liu Y, Cui B, Zhao Y. Androgen receptor gene methylation related to colorectal cancer risk. Endocr Connect 2019; 8:979-987. [PMID: 31176300 PMCID: PMC6612065 DOI: 10.1530/ec-19-0122] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 06/06/2019] [Indexed: 12/14/2022]
Abstract
According to its incidence patterns, colorectal cancer (CRC) tends to occur more frequently in males than in females, and the evidence shows that CRC is a hormone-related tumor. These findings indicate that androgen receptor (AR) gene methylation might be important for the regulation of the CRC risk in the different sexes. We used a case-control study to investigate the association between AR methylation in peripheral blood (PBL) and CRC risk. A cohort study was conducted to analyze the effect of AR methylation levels in both PBL and tissue on the prognosis of CRC. AR methylation levels were detected using methylation-sensitive high-resolution melting (MS-HRM). The results indicate that the hypomethylation of AR was significantly associated with the risk of CRC (OR = 1.869, 95% CI: 1.629-2.141, P < 0.001), and the results remained similar after adjusting for the propensity score (PS) (OR = 1.344, 95% CI: 1.147-1.575, P < 0.001) and PS matching (OR = 1.138, 95% CI: 1.000-1.292 P = 0.049). The hypomethylation of AR was significantly associated with CRC in males (OR = 2.309, 95% CI: 1.200-4.245; P = 0.012) but not females (OR = 1.000, 95% CI: 0.567-1.765; P = 0.999). The methylation status of AR in PBL and tissue does not seem to be associated with prognosis in colorectal cancer (OR = 1.425, 95% CI: 0.895-2.269, P = 0.135; OR = 0.930, 95% CI: 0.674-1.285, P = 0.661). We conclude that AR hypomethylation in PBL is associated with a high risk of CRC and may serve as a biomarker. Further studies involving large sample sizes are needed to validate the results of this study.
Collapse
Affiliation(s)
- Tingting Xia
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Hongru Sun
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Hao Huang
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Haoran Bi
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Rui Pu
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Lei Zhang
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Yuanyuan Zhang
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Ying Liu
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Jing Xu
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Justina Ucheojor Onwuka
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Yupeng Liu
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
- Correspondence should be addressed to Y Liu or B Cui or Y Zhao: or or
| | - Binbin Cui
- Department of Colorectal Surgery, The Third Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
- Correspondence should be addressed to Y Liu or B Cui or Y Zhao: or or
| | - Yashuang Zhao
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
- Correspondence should be addressed to Y Liu or B Cui or Y Zhao: or or
| |
Collapse
|
15011
|
Nesiu A, Cimpean AM, Ceausu RA, Adile A, Ioiart I, Porta C, Mazzanti M, Camerota TC, Raica M. Intracellular Chloride Ion Channel Protein-1 Expression in Clear Cell Renal Cell Carcinoma. Cancer Genomics Proteomics 2019; 16:299-307. [PMID: 31243111 PMCID: PMC6609261 DOI: 10.21873/cgp.20135] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND/AIM Chloride intracellular channel 1 (CLIC1) represents a promising target for personalized therapy. Our aim was to assess CLIC1 expression in clear cell renal cell carcinoma (cc RCC) and identify its possible prognostic role. MATERIALS AND METHODS Fifty cases of cc RCC were evaluated and selected for immunohistochemistry. CLIC1 expression was correlated with tumor grade, invasion and heterogeneity. RESULTS A total of 87.5% of the cases were CLIC1 positive, with either a homogeneous (31.42%) or a heterogeneous (68.57%) pattern. Low, mild and strong CLIC1 expressing tumors were defined based on nuclear (N), cytoplasmic (C), membrane (M) or combinations of them (NC, NM, CM, NCM) in terms of CLIC1 distribution. A significant correlation was found between tumor grade and percent of positive tumor cells (p=0.017). For G3 tumors, CLIC1 cytoplasmic expression was strongly correlated with high expression status (p=0.025) and tumor heterogeneity (p=0.004). CLIC1 expression was also correlated with metastasis (p=0.046). CONCLUSION We defined four cc RCC groups depending on G, CLIC1 expression and pattern: i) G3/NM/low CLIC1+, ii) G2/CM/mild CLIC1+ iii) G1 or G2/NM or CM /high CLIC1+, and iv) G2/M /high CLIC1.
Collapse
Affiliation(s)
- Alexandru Nesiu
- Department of Urology, Vasile Goldis University, Arad, Romania
| | - Anca Maria Cimpean
- Department of Microscopic Morphology/Histology, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
- Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Raluca Amalia Ceausu
- Department of Microscopic Morphology/Histology, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
- Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Ahmed Adile
- Department of Urology, Vasile Goldis University, Arad, Romania
| | - Ioan Ioiart
- Department of Urology, Vasile Goldis University, Arad, Romania
| | - Camillo Porta
- Department of Internal Medicine, University of Pavia & Division of Translational Oncology, IRCCS ICS Maugeri of Pavia, Pavia, Italy
| | - Michele Mazzanti
- Department of Biosciences, Laboratory of Cellular and Molecular Physiology, University of Milano, Milan, Italy
| | | | - Marius Raica
- Department of Microscopic Morphology/Histology, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
- Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| |
Collapse
|
15012
|
Kao YH, Lin WT, Thomas CL, Lin HY, Tseng TS. Association between smoking and neutrophil to lymphocyte ratio among prostate cancer survivors: the National Health and Nutrition Examination Survey. Transl Cancer Res 2019; 8:S346-S354. [PMID: 35117112 PMCID: PMC8799291 DOI: 10.21037/tcr.2019.05.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/10/2019] [Indexed: 12/02/2022]
Abstract
BACKGROUND An elevated neutrophil to lymphocyte ratio (NLR) has been proven as a vital predictor of progression and mortality for prostate cancer. Smoking is a risk factor for increasing NLR. The objective of this study is to examine the effect of smoking on NLR among prostate cancer survivors. METHODS A total of 354 men adults aged ≥20 years old with prostate cancer were analyzed using the National Health and Nutrition Examination Survey (NHANES) 2005-2016 data. The primary outcome was NLR, which was classified into two levels: high (≥3) and low (<3). Sampling weighted logistic regressions were used for evaluating associations between smoking and NLR among prostate cancer survivors. RESULTS This study found that 33.2% (n=111) of prostate cancer survivors had an elevated NLR. Prostate cancer survivors with a high NLR were older (mean 73.5 years old), non-Hispanic white (38.5%), higher income (poverty income ratio >1, 34.7%), and longer years after diagnosis (8.8 years) compared to counterparts with a low NLR. Smoking status did not have a significant impact on NLR. The interaction test between race and smoking status was significant (P=0.04). Non-Hispanic black who were current smokers were observed more likely to have high NLR than never smokers [adjusted odds ratio (OR) =3.69, 95% CI: 1.36-9.99]. However, the effect of smoking on NLR was not observed among either non-Hispanic whites or other races. CONCLUSIONS Non-Hispanic black prostate cancer survivors who were a current smoker were more likely to have NLR ≥3 compared to non-smokers. Smoking cessation could benefit these patients in prostate cancer management and reduce the risk of progression and mortality.
Collapse
Affiliation(s)
- Yu-Hsiang Kao
- Behavioral and Community Health Sciences, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Wei-Ting Lin
- Department of Global Community Health and Behavioral Sciences, Tulane University School of Public Health Tropical Medicine, New Orleans, LA, USA
| | - Casey L. Thomas
- Behavioral and Community Health Sciences, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Hui-Yi Lin
- Department of Biostatistics, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Tung-Sung Tseng
- Behavioral and Community Health Sciences, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
15013
|
Guan W, Wang X, Lin Q, Zhang J, Ren W, Xu G. Transforming growth factor‑β/miR‑143‑3p/cystatin B axis is a therapeutic target in human ovarian cancer. Int J Oncol 2019; 55:267-276. [PMID: 31180557 DOI: 10.3892/ijo.2019.4815] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 05/24/2019] [Indexed: 12/24/2022] Open
Abstract
We previously reported that cystatin B (CSTB) is a progression marker of human ovarian cancer (OC); however, the regulatory mechanism of CSTB and its function in OC remain unclear. The present study aimed to explore the mechanism underlying transforming growth factor-β (TGF‑β) 1‑mediated CSTB regulation, and to examine the function of CSTB on OC cell proliferation and apoptosis. Using the online program, miRWalk, a microRNA (miR)‑143‑3p was detected, which contains a homologous sequence of the potential binding site to the 3'‑untranslated region (3'‑UTR) of CSTB. A dual‑luciferase reporter assay confirmed the interaction between miR‑143‑3p and CSTB 3'‑UTR. Treating OC cells with miR‑143‑3p mimics or inhibitors resulted in a decrease or an increase of CSTB expression at mRNA and protein levels, respectively. Additionally, CSTB was significantly overexpressed, whereas miR‑143‑3p was downregulated in human OC tissues compared with normal ovarian tissues. A negative correlation between miR‑143‑3p and CSTB mRNA expression was observed in ovarian malignant tumors. The levels of primary and mature miR‑143‑3p expression were upregulated in OC cells after TGF‑β1 treatment; the action of TGF‑β1 was abolished in the presence of an inhibitor of TGF‑β type I receptor. These results indicated an axis between TGF‑β, miR‑143‑3p and CSTB in OC cells. Furthermore, high levels of CSTB expression were associated with the poor overall survival of patients with OC. Knockdown of CSTB resulted in a decrease in OC cell proliferation and arrested cells in G2/M phase. In addition, suppression of CSTB induced cell apoptosis. In conclusion, CSTB was overexpressed and miR‑143‑3p was downregulated in ovarian malignant tumors. Mature miR‑143‑3p directly bound CSTB 3'‑UTR, leading to a decrease in CSTB expression in OC cells, which was regulated by TGF‑β1. Our findings suggest the potential therapeutic application of targeting the TGF‑β/miR‑143‑3p/CSTB axis for treating patients with OC.
Collapse
Affiliation(s)
- Wencai Guan
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Xingxing Wang
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Qunbo Lin
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Jinguo Zhang
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Weimin Ren
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Guoxiong Xu
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| |
Collapse
|
15014
|
Wu L, Wang J, Cai Q, Cavazos TB, Emami NC, Long J, Shu XO, Lu Y, Guo X, Bauer JA, Pasaniuc B, Penney KL, Freedman ML, Kote-Jarai Z, Witte JS, Haiman CA, Eeles RA, Zheng W. Identification of Novel Susceptibility Loci and Genes for Prostate Cancer Risk: A Transcriptome-Wide Association Study in Over 140,000 European Descendants. Cancer Res 2019; 79:3192-3204. [PMID: 31101764 PMCID: PMC6606384 DOI: 10.1158/0008-5472.can-18-3536] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/04/2019] [Accepted: 05/09/2019] [Indexed: 11/16/2022]
Abstract
Genome-wide association study-identified prostate cancer risk variants explain only a relatively small fraction of its familial relative risk, and the genes responsible for many of these identified associations remain unknown. To discover novel prostate cancer genetic loci and possible causal genes at previously identified risk loci, we performed a transcriptome-wide association study in 79,194 cases and 61,112 controls of European ancestry. Using data from the Genotype-Tissue Expression Project, we established genetic models to predict gene expression across the transcriptome for both prostate models and cross-tissue models and evaluated model performance using two independent datasets. We identified significant associations for 137 genes at P < 2.61 × 10-6, a Bonferroni-corrected threshold, including nine genes that remained significant at P < 2.61 × 10-6 after adjusting for all known prostate cancer risk variants in nearby regions. Of the 128 remaining associated genes, 94 have not yet been reported as potential target genes at known loci. We silenced 14 genes and many showed a consistent effect on viability and colony-forming efficiency in three cell lines. Our study provides substantial new information to advance our understanding of prostate cancer genetics and biology. SIGNIFICANCE: This study identifies novel prostate cancer genetic loci and possible causal genes, advancing our understanding of the molecular mechanisms that drive prostate cancer.
Collapse
Affiliation(s)
- Lang Wu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, Hawaii
| | - Jifeng Wang
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Urology, The Fifth People's Hospital of Shanghai, Shanghai, China
| | - Qiuyin Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Taylor B Cavazos
- Program in Biological and Medical Informatics, University of California, San Francisco, San Francisco, California
| | - Nima C Emami
- Program in Biological and Medical Informatics, University of California, San Francisco, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yingchang Lu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Xingyi Guo
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joshua A Bauer
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee
- Vanderbilt Institute of Chemical Biology, High-Throughput Screening Facility, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Bogdan Pasaniuc
- Department of Pathology and Laboratory Medicine and Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Kathryn L Penney
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | | | - Zsofia Kote-Jarai
- Division of Genetics and Epidemiology, The Institute of Cancer Research, and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - John S Witte
- Program in Biological and Medical Informatics, University of California, San Francisco, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Christopher A Haiman
- Department of Preventive Medicine, University of Southern California, Los Angeles, California
| | - Rosalind A Eeles
- Division of Genetics and Epidemiology, The Institute of Cancer Research, and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
15015
|
Fowler AM. Survival Outcomes for Women with Ductal Carcinoma in Situ in the Era of Supplemental Screening. Radiology 2019; 292:49-50. [DOI: 10.1148/radiol.2019190762] [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)
- Amy M. Fowler
- From the Department of Radiology, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, Madison, WI 53792-3252; University of Wisconsin Carbone Cancer Center, Madison, Wis; and Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| |
Collapse
|
15016
|
Nam S, Khawar IA, Park JK, Chang S, Kuh HJ. Cellular context-dependent interaction between cancer and stellate cells in hetero-type multicellular spheroids of pancreatic tumor. Biochem Biophys Res Commun 2019; 515:183-189. [DOI: 10.1016/j.bbrc.2019.05.101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 05/13/2019] [Indexed: 11/24/2022]
|
15017
|
Magnani CJ, Li K, Seto T, McDonald KM, Blayney DW, Brooks JD, Hernandez-Boussard T. PSA Testing Use and Prostate Cancer Diagnostic Stage After the 2012 U.S. Preventive Services Task Force Guideline Changes. J Natl Compr Canc Netw 2019; 17:795-803. [PMID: 31319390 PMCID: PMC7195904 DOI: 10.6004/jnccn.2018.7274] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 01/15/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Most patients with prostate cancer are diagnosed with low-grade, localized disease and may not require definitive treatment. In 2012, the U.S. Preventive Services Task Force (USPSTF) recommended against prostate cancer screening to address overdetection and overtreatment. This study sought to determine the effect of guideline changes on prostate-specific antigen (PSA) screening and initial diagnostic stage for prostate cancer. PATIENTS AND METHODS A difference-in-differences analysis was conducted to compare changes in PSA screening (exposure) relative to cholesterol testing (control) after the 2012 USPSTF guideline changes, and chi-square test was used to determine whether there was a subsequent decrease in early-stage, low-risk prostate cancer diagnoses. Data were derived from a tertiary academic medical center's electronic health records, a national commercial insurance database (OptumLabs), and the SEER database for men aged ≥35 years before (2008-2011) and after (2013-2016) the guideline changes. RESULTS In both the academic center and insurance databases, PSA testing significantly decreased for all men compared with the control. The greatest decrease was among men aged 55 to 74 years at the academic center and among those aged ≥75 years in the commercial database. The proportion of early-stage prostate cancer diagnoses ( CONCLUSIONS In primary care, PSA testing decreased significantly and fewer prostate cancers were diagnosed at an early stage, suggesting provider adherence to the 2012 USPSTF guideline changes. Long-term follow-up is needed to understand the effect of decreased screening on prostate cancer survival.
Collapse
Affiliation(s)
| | - Kevin Li
- School of Medicine, Stanford University
| | - Tina Seto
- Stanford University School of Medicine IRT Research Technology
| | | | - Douglas W. Blayney
- Department of Medicine, Stanford University
- Stanford Cancer Institute, Stanford University
| | | | - Tina Hernandez-Boussard
- Department of Medicine, Stanford University
- Department of Biomedical Data Science, Stanford University
| |
Collapse
|
15018
|
Ajani JA, D'Amico TA, Bentrem DJ, Chao J, Corvera C, Das P, Denlinger CS, Enzinger PC, Fanta P, Farjah F, Gerdes H, Gibson M, Glasgow RE, Hayman JA, Hochwald S, Hofstetter WL, Ilson DH, Jaroszewski D, Johung KL, Keswani RN, Kleinberg LR, Leong S, Ly QP, Matkowskyj KA, McNamara M, Mulcahy MF, Paluri RK, Park H, Perry KA, Pimiento J, Poultsides GA, Roses R, Strong VE, Wiesner G, Willett CG, Wright CD, McMillian NR, Pluchino LA. Esophageal and Esophagogastric Junction Cancers, Version 2.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2019; 17:855-883. [PMID: 31319389 DOI: 10.6004/jnccn.2019.0033] [Citation(s) in RCA: 655] [Impact Index Per Article: 109.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Esophageal cancer is the sixth leading cause of cancer-related deaths worldwide. Squamous cell carcinoma is the most common histology in Eastern Europe and Asia, and adenocarcinoma is most common in North America and Western Europe. Surgery is a major component of treatment of locally advanced resectable esophageal and esophagogastric junction (EGJ) cancer, and randomized trials have shown that the addition of preoperative chemoradiation or perioperative chemotherapy to surgery significantly improves survival. Targeted therapies including trastuzumab, ramucirumab, and pembrolizumab have produced encouraging results in the treatment of patients with advanced or metastatic disease. Multidisciplinary team management is essential for all patients with esophageal and EGJ cancers. This selection from the NCCN Guidelines for Esophageal and Esophagogastric Junction Cancers focuses on recommendations for the management of locally advanced and metastatic adenocarcinoma of the esophagus and EGJ.
Collapse
Affiliation(s)
| | | | - David J Bentrem
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | - Prajnan Das
- The University of Texas MD Anderson Cancer Center
| | | | | | | | - Farhood Farjah
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | | | | | | | | | | | | | | | | | - Rajesh N Keswani
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | | | | | - Michael McNamara
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | - Mary F Mulcahy
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | - Haeseong Park
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | - Kyle A Perry
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | | | | | - Robert Roses
- Abramson Cancer Center at the University of Pennsylvania
| | | | | | | | | | | | | |
Collapse
|
15019
|
|
15020
|
Lin HY, Fisher P, Harris D, Tseng TS. Alcohol intake patterns for cancer and non-cancer individuals: a population study. Transl Cancer Res 2019; 8:S334-S345. [PMID: 31497514 PMCID: PMC6731030 DOI: 10.21037/tcr.2019.06.31] [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] [Indexed: 12/20/2022]
Abstract
Background Alcohol intake is a leading modifiable cause related to cancer-specific deaths. Various alcohol intake patterns have shown to impact cancer progression differently, however, many studies only evaluated simplified patterns (such as heavy vs. non-heavy drinking) of alcohol intake for cancer survivors. The objective of this study is to provide population-based prevalence of the complex alcohol drinking patterns for cancer survivors, and compare it with that of non-cancer individuals. Additionally, we evaluated the impact of cancer related factors (binary, alcohol-related cancer type, and length of cancer history) to the alcohol intake patterns adjusted for the selected factors. Methods A total of 193,197 individuals, including 16,504 cancer survivors, with age ≥18 years old in the 2012–2017 National Health Interview Survey (NHIS) were included in this study. The population-based prevalence of alcohol patterns was estimated. To evaluate cancer related factors associated with the alcohol intake patterns, we applied multinomial logistic models with the appropriate sampling weights and adjusted the selected demographic factors and smoking status. Results There were 62.1% of cancer survivors and 66.0% of non-cancer individuals who were current alcohol drinkers in the past year. The prevalence of heavy drinking was identical for 5.2% of cancer and non-cancer individuals. For frequent binge drinking, cancer survivors tended to have less frequent binge than non-cancer individuals (2.8% vs. 4.9%). After adjusting for the selected demographic factors and smoking status, the cancer survivors were less likely to have the intermediate level of alcohol intake (light/moderate or occasional binge drinking) compared with non-cancer individuals, but no difference for the excessive alcohol intake (heavy or frequent binge drinking) was observed for those with and without cancer. As for cancer type, those with non-alcohol related cancer tended to be a current drinker compared with those with alcohol-related cancer. Compared with cancer survivors with a short cancer history (2–4 years), survivors with a cancer history of 5–9 years were more likely to be current drinkers after adjusting for the selected factors. Cancer status, alcohol-related cancer type and length of cancer history had no impact on excessive alcohol intake. Conclusions In summary, cancer survivors have similar excessive alcohol drinking patterns but were less likely to have the intermediate level of alcohol intake compared to non-cancer individuals. Alcohol intake may enhance cancer progression, interfere with cancer treatments and increase cancer-related mortality. To improve cancer survivors’ health, custom alcohol interventions and cessation programs should be conducted to minimize alcohol intake for cancer survivors, especially for excessive alcohol drinkers.
Collapse
Affiliation(s)
- Hui-Yi Lin
- School of Public Health, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Paige Fisher
- School of Public Health, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Darian Harris
- School of Public Health, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Tung-Sung Tseng
- School of Public Health, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| |
Collapse
|
15021
|
Liu Y, Sun H, Makabel B, Cui Q, Li J, Su C, Ashby CR, Chen Z, Zhang J. The targeting of non‑coding RNAs by curcumin: Facts and hopes for cancer therapy (Review). Oncol Rep 2019; 42:20-34. [PMID: 31059075 PMCID: PMC6549103 DOI: 10.3892/or.2019.7148] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 04/24/2019] [Indexed: 02/07/2023] Open
Abstract
Curcumin [(1E,6E)‑1,7‑bis(4‑hydroxy‑3‑-methoxyphenyl) hepta‑1,6‑diene‑3,5‑dione] is a natural polyphenol that is derived from the turmeric plant (curcuma longa L.). Curcumin is widely used in food coloring, preservatives, and condiments. Curcumin possesses anti‑tumor, anti‑oxidative and anti‑inflammatory efficacy, as well as other pharmacological effects. Emerging evidence indicates that curcumin alters microRNAs (miRNAs) and long non‑coding RNAs (lncRNAs) in various types of cancers. Both miRNAs and lncRNAs are non‑coding RNAs that can epigenetically modulate the expression of multiple genes via post‑transcriptional regulation. In the present review, the interactions between curcumin and non‑coding RNAs are summarized in numerous types of cancers, including lung, colorectal, prostate, breast, nasopharyngeal, pancreatic, blood, and ovarian cancer, and the vital non‑coding RNAs and their downstream targets are described.
Collapse
Affiliation(s)
- Yun Liu
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Hongmei Sun
- Infinitus (China) Company Ltd., Jiangmen, Guangdong 529156, P.R. China
| | - Bolat Makabel
- Xinjiang Institute of Materia Medica, Urumqi, Xinjiang 830004, P.R. China
| | - Qingbin Cui
- College of Pharmacy and Health Sciences, St. John's University, Queens, New York, NY 11439, USA
- College of Public Health, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Jiajun Li
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Chaoyue Su
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Charles R. Ashby
- College of Pharmacy and Health Sciences, St. John's University, Queens, New York, NY 11439, USA
| | - Zhesheng Chen
- College of Pharmacy and Health Sciences, St. John's University, Queens, New York, NY 11439, USA
| | - Jianye Zhang
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| |
Collapse
|
15022
|
Nguyen C, Pandey S. Exploiting Mitochondrial Vulnerabilities to Trigger Apoptosis Selectively in Cancer Cells. Cancers (Basel) 2019; 11:E916. [PMID: 31261935 PMCID: PMC6678564 DOI: 10.3390/cancers11070916] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/19/2019] [Accepted: 06/25/2019] [Indexed: 12/14/2022] Open
Abstract
The transformation of normal cells to the cancerous stage involves multiple genetic changes or mutations leading to hyperproliferation, resistance to apoptosis, and evasion of the host immune system. However, to accomplish hyperproliferation, cancer cells undergo profound metabolic reprogramming including oxidative glycolysis and acidification of the cytoplasm, leading to hyperpolarization of the mitochondrial membrane. The majority of drug development research in the past has focused on targeting DNA replication, repair, and tubulin polymerization to induce apoptosis in cancer cells. Unfortunately, these are not cancer-selective targets. Recently, researchers have started focusing on metabolic, mitochondrial, and oxidative stress vulnerabilities of cancer cells that can be exploited as selective targets for inducing cancer cell death. Indeed, the hyperpolarization of mitochondrial membranes in cancer cells can lead to selective importing of mitocans that can induce apoptotic effects. Herein, we will discuss recent mitochondrial-selective anticancer compounds (mitocans) that have shown selective toxicity against cancer cells. Increased oxidative stress has also been shown to be very effective in selectively inducing cell death in cancer cells. This oxidative stress could lead to mitochondrial dysfunction, which in turn will produce more reactive oxygen species (ROS). This creates a vicious cycle of mitochondrial dysfunction and ROS production, irreversibly leading to cell suicide. We will also explore the possibility of combining these compounds to sensitize cancer cells to the conventional anticancer agents. Mitocans in combination with selective oxidative-stress producing agents could be very effective anticancer treatments with minimal effect on healthy cells.
Collapse
Affiliation(s)
- Christopher Nguyen
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON N9E 3P4, Canada
| | - Siyaram Pandey
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON N9E 3P4, Canada.
| |
Collapse
|
15023
|
Feng FM, Zhang L. Analysis of KLF5 expression and its prognostic significance in gastric cancer based on Oncomine and Kaplan-Meier Plotter. Shijie Huaren Xiaohua Zazhi 2019; 27:734-741. [DOI: 10.11569/wcjd.v27.i12.734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is a common malignancy of the digestive system. Most patients with advanced GC have a poor prognosis and lack of prognostic predictors.
AIM To analyze the expression of Kruppel-like factor 5 (KLF5) in GC and its correlation with patient prognosis by using Oncomie and Kaplan-Meier Plotter databases.
METHODS The data sets of KLF5 gene expression in Oncomie and Kaplan-Meier Plotter databases were collected. The differential expression of KLF5 gene in GC tissues was deeply mined. According to the median expression level of KLF5, the patients were divided into a high-expression group and low-expression group. Survival curves were drawn and log-rank test was used to compare the overall survival and disease-free survival of the two groups. Meanwhile, the clinical data of 41 patients with GC treated at our hospital were retrospectively analyzed. The expression of KLF5 in the 41 patients with GC was detected by immunohistochemistry, and the relationship between the expression of KLF5 and the clinicopathological characteristics of patients was analyzed.
RESULTS A total of 424 studies on KLF5 gene expression were collected in Oncomine database, of which 35 showed differential expression of KLF5 in normal vs cancer tissues. Compared with normal tissues, 8 datasets showed that KLF5 was highly expressed in cancer tissues and 27 datasets showed low expression in cancer tissues. Cluster analysis showed that KLF5 was co-expressed with 20 genes such as ST14 and TMEM125 (co-down-regulated or up-regulated), suggesting that these co-expressed genes might be functionally related. The differential expression of KLF5 gene in GC tissues was analyzed by using 10 data sets of KLF5 expression chips from Oncomine database. Four of them indicated that the expression level of KLF5 in GC tissues was significantly increased. In the Kaplan-Meier Plotter database, two related gene chips were used to analyze the relationship between the expression of KLF5 and the prognosis of GC patients. The long-rank test showed that the overall survival and disease-free survival of patients with high expression of KLF5 were less than those of patients with low expression (P < 0.05). Immunohistochemical analysis showed that 29 (70.7%) of 41 patients with GC were KLF5 positive. There was no significant correlation between KLF5 positive expression and clinicopathological features such as gender, age, tumor stage, or tumor grade (P > 0.05).
CONCLUSION The high expression of KLF5 in GC is associated with a poor prognosis, although the expression of KLF5 is not related to the clinicopathological characteristics of GC patients.
Collapse
Affiliation(s)
- Fu-Mei Feng
- Department of Gastrointestinal Surgery, Tianjin Baodi District People's Hospital, Tianjin 301800, China
| | - Lei Zhang
- Department of Gastrointestinal Surgery, Tianjin Baodi District People's Hospital, Tianjin 301800, China
| |
Collapse
|
15024
|
Basuli F, Zhang X, Blackman B, White ME, Jagoda EM, Choyke PL, Swenson RE. Fluorine-18 Labeled Fluorofuranylnorprogesterone ([ 18F]FFNP) and Dihydrotestosterone ([ 18F]FDHT) Prepared by "Fluorination on Sep-Pak" Method. Molecules 2019; 24:molecules24132389. [PMID: 31261651 PMCID: PMC6651117 DOI: 10.3390/molecules24132389] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 06/25/2019] [Accepted: 06/25/2019] [Indexed: 12/31/2022] Open
Abstract
To further explore the scope of our recently developed “fluorination on Sep-Pak” method, we prepared two well-known positron emission tomography (PET) tracers 21-[18F]fluoro-16α,17α-[(R)-(1′-α-furylmethylidene)dioxy]-19-norpregn-4-ene-3,20-dione furanyl norprogesterone ([18F]FFNP) and 16β-[18F]fluoro-5α-dihydrotestosterone ([18F]FDHT). Following the “fluorination on Sep-Pak” method, over 70% elution efficiency was observed with 3 mg of triflate precursor of [18F]FFNP. The overall yield of [18F]FFNP was 64–72% (decay corrected) in 40 min synthesis time with a molar activity of 37–81 GBq/µmol (1000–2200 Ci/mmol). Slightly lower elution efficiency (~55%) was observed with the triflate precursor of [18F]FDHT. Fluorine-18 labeling, reduction, and deprotection to prepare [18F]FDHT were performed on Sep-Pak cartridges (PS-HCO3 and Sep-Pak plus C-18). The overall yield of [18F]FDHT was 25–32% (decay corrected) in 70 min. The molar activity determined by using mass spectrometry was 63–148 GBq/µmol (1700–4000 Ci/mmol). Applying this quantitative measure of molar activity to in vitro assays [18F]FDHT exhibited high-affinity binding to androgen receptors (Kd~2.5 nM) providing biological validation of this method.
Collapse
Affiliation(s)
- Falguni Basuli
- Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD 20850, USA.
| | - Xiang Zhang
- Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD 20850, USA
| | - Burchelle Blackman
- Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD 20850, USA
| | - Margaret E White
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elaine M Jagoda
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter L Choyke
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rolf E Swenson
- Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD 20850, USA
| |
Collapse
|
15025
|
Long noncoding RNAs as potential biomarkers and therapeutic targets in gallbladder cancer: a systematic review and meta-analysis. Cancer Cell Int 2019; 19:169. [PMID: 31297033 PMCID: PMC6599267 DOI: 10.1186/s12935-019-0891-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/23/2019] [Indexed: 02/06/2023] Open
Abstract
Background Mounting evidence has shown that long noncoding RNAs (lncRNAs) can play a substantial role in gallbladder cancer (GBC) development as tumor promotors or suppressors, and their abnormal expression is relevant to GBC patient outcomes. We completed this systematic review and meta-analysis to explore the clinical significance and mechanisms of lncRNAs in GBC. Methods We conducted a comprehensive literature search and selected eligible records according to the inclusion and exclusion criteria. Hazard ratios (HRs) and odds ratios (ORs) were extracted or calculated to estimate the relationships of high lncRNA expression with GBC patient survival and clinical outcomes. Results Eighteen studies were identified as eligible for this systematic review and meta-analysis. Heterogeneity among HRs of overall survival (OS) was notably high (I2 = 86.2%, p < 0.001). Subgroup analysis suggested that overexpression of lncRNAs in a group that is upregulated in GBC showed a significant association with poor OS (HR = 2.454, 95% CI 2.004–3.004, I2 = 0%). Conversely, overexpression of lncRNAs in a downregulated group was markedly related to good OS (HR = 0.371, 95% CI 0.267–0.517, I2 = 0%). High expression levels of lncRNA AFAP1-AS1, MALAT1 and ROR were positively correlated with tumor size. Expression of lncRNA LET, LINC00152 and HEGBC exhibited a positive correlation with high T status. LncRNA LINC00152, HEGBC, MALAT1 and ROR showed a marked correlation with positive lymph node metastasis (LNM), while lncRNA GCASPC, MEG3, LET and UCA1 had the opposite effect. High expression levels of lncRNA HEGBC, PAGBC, PVT1 and UCA1 predicted high tumor node metastasis (TNM) stages, while lncRNA LET, GCASPC and MEG3 indicated low TNM stages. We also summarized the mechanisms of lncRNAs in GBC. Conclusion Aberrant expression of several lncRNAs was indicative of the prognosis of GBC patients, and lncRNAs showed promise as biomarkers and therapeutic targets for GBC.
Collapse
|
15026
|
Caveolin-1 enhances brain metastasis of non-small cell lung cancer, potentially in association with the epithelial-mesenchymal transition marker SNAIL. Cancer Cell Int 2019; 19:171. [PMID: 31297035 PMCID: PMC6599320 DOI: 10.1186/s12935-019-0892-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 06/25/2019] [Indexed: 02/08/2023] Open
Abstract
Background Caveolin-1 (Cav-1) plays an important role in the development of various human cancers. We investigated the relationship between Cav-1 expression and non-small cell lung cancer (NSCLC) progression in the context of brain metastasis (BM). Methods Cav-1 expression was investigated in a series of 102 BM samples and 49 paired primary NSCLC samples, as well as 162 unpaired primary NSCLC samples with (63 cases) or without (99 cases) metastasis to distant organs. Human lung cancer cell lines were used for in vitro functional analysis. Results High Cav-1 expression in tumor cells was observed in 52% (38/73) of squamous cell carcinomas (SQCs) and 33% (45/138) of non-SQCs. In SQC, high Cav-1 expression was increased after BM in both paired and unpaired samples of lung primary tumors and BM (53% vs. 84% in paired samples, P = 0.034; 52% vs. 78% in unpaired samples, P = 0.020). Although the difference in median overall survival in patients NSCLC was not statistically significant, high Cav-1 expression in tumor cells (P = 0.005, hazard ratio 1.715, 95% confidence index 1.175–2.502) was independent prognostic factors of overall survival on multivariate Cox regression analyses, in addition to the presence of BM and non-SQC type. In vitro assays revealed that Cav-1 knockdown inhibited the invasion and migration of lung cancer cells. Genetic modulation of Cav-1 was consistently associated with SNAIL up- and down-regulation. These findings were supported by increased SNAIL and Cav-1 expression in BM samples of SQC. Conclusions Cav-1 plays an important role in the BM of NSCLC, especially in SQC. The mechanism may be linked to SNAIL regulation. Electronic supplementary material The online version of this article (10.1186/s12935-019-0892-0) contains supplementary material, which is available to authorized users.
Collapse
|
15027
|
Li J, Chen L, Yan L, Gu Z, Chen Z, Zhang A, Zhao F. A Novel Drug Design Strategy: An Inspiration from Encaging Tumor by Metallofullerenol Gd@C 82(OH) 22. Molecules 2019; 24:molecules24132387. [PMID: 31252662 PMCID: PMC6650816 DOI: 10.3390/molecules24132387] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 06/24/2019] [Accepted: 06/26/2019] [Indexed: 12/30/2022] Open
Abstract
Cancer remains a major threat to human health worldwide. Cytotoxicity has imposed restrictions on the conventional cytotoxic drug-based chemotherapy. The rapidly-developing nanomedicine has shown great promise in revolutionizing chemotherapy with improved efficiency and reduced toxicity. Gd@C82(OH)22, a novel endohedral metallofullerenol, was first reported by our research group to suppress tumor growth and metastasis efficiently without obvious toxicity. Gd@C82(OH)22 imprisons tumors by facilitating the formation of surrounding fibrous layers which is different from chemotherapeutics that poison tumor cells. In this review, the authors first reported the antineoplastic activity of metallofullerenol Gd@C82(OH)22 followed by further discussions on its new anti-cancer molecular mechanism—tumor encaging. On this basis, the unparalleled advantages of nanomedicine in the future drug design are discussed. The unique interaction modes of Gd@C82(OH)22 with specific targeted biomolecules may shed light on a new avenue for drug design. Depending on the surface characteristics of target biomolecules, nanomedicine, just like a transformable and dynamic key, can self-assemble into suitable shapes to match several locks for the thermodynamic stability, suggesting the target-tailoring ability of nanomedicine.
Collapse
Affiliation(s)
- Jinxia Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Linlin Chen
- College of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Liang Yan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Zhanjun Gu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Zhaofang Chen
- Jiangsu Key Laboratory of Hazardous Chemicals Safety and Control, College of Safety Science and Engineering, Nanjing Tech University, Nanjing 210009, China
| | - Aiping Zhang
- College of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Feng Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China.
| |
Collapse
|
15028
|
Zheng W, Zhang Y, Guo L, Wang S, Fang M, Mao W, Lou J. Evaluation of therapeutic efficacy with CytoSorter ® circulating tumor cell-capture system in patients with locally advanced head and neck squamous cell carcinoma. Cancer Manag Res 2019; 11:5857-5869. [PMID: 31303792 PMCID: PMC6603285 DOI: 10.2147/cmar.s208409] [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: 03/12/2019] [Accepted: 05/21/2019] [Indexed: 01/20/2023] Open
Abstract
Objective: This study aimed to investigate the feasibility of utilizing CytoSorter® system to detect circulating tumor cells (CTCs) and clinical value of CTCs in patients with locally advanced head and neck squamous cell carcinoma (LAHNSCC). Methods: 31 patients with LAHNSCC, 12 healthy volunteers, and 6 patients with benign tumor serving as controls were enrolled in this study. CTCs were enriched with the CytoSorter®, a microfluidic based immune capture system. CTC detection was performed before and after induction chemotherapy, as well as after surgery and/or radiotherapy. Correlations between CTC enumerations at different time points and survival outcome and recurrence risk were evaluated. The correlation between CTCs and clinicopathological characteristics was appraised. Follow-up of patients continued until March 2019. Results: While CTCs were not found in the controls, they were detected in 24 of 31 LAHNSCC patients. CTCs could be used to distinguish diseased people from the healthy (P<0.0001). CTCs were statistically associated with patient age (P=0.037, >60 years old vs<60 years old) and lymph node metastasis (P= 0.034, N0N1 VS N2N3). Most patients had significantly reduced CTCs at the end of treatment. Patients with partial remission of tumor after induction therapy had more CTCs than those with complete remission of tumor. Patients with higher CTCs counts prior to treatment had higher chance of developing local recurrence of tumor after treatment (P=0.0187). Conclusion: CTCs were successfully isolated in LAHNSCC patients using CytoSorter® system with better sensibility. CTCs can be used to differentiate LAHNSCC patients from those with benign HNSCC tumor or healthy volunteers, and as markers to monitor patient’s response to treatment and predict the local tumor recurrence after treatment. CTC detection at baseline has the greatest prognostic potency in LAHNSCC patients.
Collapse
Affiliation(s)
- Weihui Zheng
- Center of Oncology, the First Affiliated Hospital of Soochow University, Suzhou 215000, People's Republic of China.,Department of Head and Neck Surgery, Zhejiang Cancer Hospital & Creative Laboratory of Head and Neck Oncology in Zhejiang Province, Hangzhou 310022, People's Republic of China
| | - Yibiao Zhang
- Department of Clinical Laboratory, Jinhua Guangfu Hospital, Jinhua 321000, People's Republic of China
| | - Liang Guo
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital & Creative Laboratory of Head and Neck Oncology in Zhejiang Province, Hangzhou 310022, People's Republic of China
| | - Shengye Wang
- Department of Radiotherapy, Zhejiang Cancer Hospital, Hangzhou 310022, People's Republic of China
| | - Meiyu Fang
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, People's Republic of China
| | - Weimin Mao
- Center of Oncology, the First Affiliated Hospital of Soochow University, Suzhou 215000, People's Republic of China
| | - Jianlin Lou
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital & Creative Laboratory of Head and Neck Oncology in Zhejiang Province, Hangzhou 310022, People's Republic of China
| |
Collapse
|
15029
|
Zhang X, Cheng D, Liu Y, Wu Y, He Z. Gephyrin suppresses lung squamous cell carcinoma development by reducing mTOR pathway activation. Cancer Manag Res 2019; 11:5333-5341. [PMID: 31239782 PMCID: PMC6560210 DOI: 10.2147/cmar.s204358] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 04/24/2019] [Indexed: 11/23/2022] Open
Abstract
Background: The mTOR pathway is altered in a multitude of cancers, including lung cancer; however, abnormal activation in this pathway is less common in lung adenocarcinoma (LUAD) than in lung squamous cell carcinoma (LUSC). Gephyrin is a highly conserved and widely expressed ancient protein in vertebrate tissues. Its role and molecular mechanism in lung cancer development are largely unknown. Method: We analyzed the expression profile of gephyrin and overall survival rates in LUAD and LUSC. The LUSC cells (H520 and SK-MES-1) were transfected with pLV-gephyrin to establish gephyrin stable overexpression cell lines. Real-time quantitative PCR and Western blot were performed to detect the mRNA and protein levels. The cell growth and cell cycle were detected by the MTT assay and flow cytometry. Finally, a xenograft tumor model was established to determine cell tumorigenesis in vivo. Results: Our results show that gephyrin was reduced in LUAD and LUSC, and its low expression in LUSC patients indicated poor prognosis. Gephyrin overexpression suppressed LUSC cell proliferation, arrested cell cycle progression, and decreased the expression of cell-cycle related proteins such as cyclin D1, cyclin-dependent kinase-2 (CDK2), and proliferation-related protein proliferating cell nuclear antigen (PCNA). Conversely, knockdown of gephyrin promoted LUSC cell growth. Moreover, gephyrin reduced mTOR pathway activation to inhibit cyclin D1 and CDK2 translation. Mechanistically, gephyrin suppressed mTOR pathway activation by promoting mTOR degradation. Furthermore, gephyrin overexpression suppressed LUSC tumorigenesis. Conclusion: Gephyrin suppressed LUSC development by reducing mTOR pathway activation, implicating gephyrin as a potential molecular target for LUSC management.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China.,Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Dezhi Cheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China.,Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Yu Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China.,Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Yuanbo Wu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Zhifeng He
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China.,Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| |
Collapse
|
15030
|
Abstract
Current recommendations by the United States Preventive Services Task Force do not support screening for skin cancer. Melanoma is unique among cancers because detection is through visual inspection. Development of technologies that aid visual inspection have supported screening strategies in high-risk populations such as older fair skinned males with personal or family history of melanoma. Clearly delineating these populations and appropriate utilization of these newer technologies will be imperative in future screening paradigms.
Collapse
Affiliation(s)
- Conor H O'Neill
- Hiram C. Polk, JR, MD Department of Surgery, Division of Surgical Oncology, University of Louisville, Louisville, Kentucky
| | - Charles R Scoggins
- Hiram C. Polk, JR, MD Department of Surgery, Division of Surgical Oncology, University of Louisville, Louisville, Kentucky
| |
Collapse
|
15031
|
Poola I, Yue Q, Gillespie JW, Sullivan PS, Aguilar-Jakthong J, Rao J, Shaaban AM, Sauter ER, Ricci AJ. Breast Hyperplasias, Risk Signature, and Breast Cancer. Cancer Prev Res (Phila) 2019; 12:471-480. [PMID: 31239263 DOI: 10.1158/1940-6207.capr-19-0051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/27/2019] [Accepted: 05/13/2019] [Indexed: 11/16/2022]
Abstract
We address the dilemma faced by oncologists in administering preventative measures to "at risk" patients diagnosed with atypical and nonatypical hyperplasias due to lack of any molecular means of risk stratification and identifying high-risk subjects. Our study purpose is to investigate a four marker risk signature, MMP-1, CEACAM6, HYAL1, and HEC1, using 440 hyperplastic tissues for identifying high-risk subjects who will benefit from preventative therapies. We assayed the markers by IHC and combined their expression levels to obtain a composite value from 0-10, which we called a "Cancer Risk Score." We demonstrate that the four marker-based risk scores predict subsequent cancer development with an accuracy of 91% and 86% for atypical and nonatypical subjects, respectively. We have established a correlation between risk scores and cancer rates by stratifying the samples into low risk (score ≤ 0.5); intermediate risk (score ≤ 5.4), and high risk (score >5.4) groups using Kaplan-Meier survival analysis. We have evaluated cancer rates at 5, 10, and 15 years. Our results show that the average cancer rates in the first 5 years among low- and intermediate-risk groups were 2% and 15%, respectively. Among high-risk group, the average cancer rates at 5 years were 73% and 34% for atypical and nonatypical subjects, respectively. The molecular risk stratification described here assesses a patient's tumor biology-based risk level as low, intermediate, or high and for making informed treatment decisions. The outcomes of our study in conjunction with the available prophylactic measures could prevent approximately 20%-25% of sporadic breast cancers.
Collapse
Affiliation(s)
| | - Qingqi Yue
- Silbiotech, Inc., Gaithersburg, Maryland
| | | | - Peggy S Sullivan
- Pathology Division, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Josephine Aguilar-Jakthong
- Pathology Division, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - JianYu Rao
- Pathology Division, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | | | - Edward R Sauter
- Department of Surgery, Hartford Hospital, Hartford, Connecticut
| | - Andrew J Ricci
- Department of Pathology, Hartford Hospital, Hartford, Connecticut
| |
Collapse
|
15032
|
Lozano MD, Abengozar-Muela M, Echeveste JI, Subtil JC, Bertó J, Gúrpide A, Calvo A, de Andrea CE. Programmed death-ligand 1 expression on direct Pap-stained cytology smears from non-small cell lung cancer: Comparison with cell blocks and surgical resection specimens. Cancer Cytopathol 2019; 127:470-480. [PMID: 31245924 DOI: 10.1002/cncy.22155] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/03/2019] [Accepted: 05/15/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) expression, as assessed by immunohistochemistry (IHC), is used to select patients with non-small cell lung cancer (NSCLC) for anti-programmed cell death protein 1 (PD-1)/PD-L1 therapy. The current study evaluated the feasibility and efficacy of PD-L1 immunostaining and quantitation on direct Papanicolaou-stained cytological smears compared with formalin-fixed paraffin-embedded samples (cytological cell blocks and surgical resection specimens) in NSCLC cases using 2 commercially available assays: the PD-L1 IHC 22C3 pharmDx assay (Agilent Technologies/Dako, Carpinteria, CA, USA) and the Ventana SP263 Assay (Ventana Medical Systems Inc, Tucson, Arizona). METHODS PD-L1 immunostaining using either both or one of the assays was tested in 117 sets of paired samples obtained from 62 NSCLC cases. The tumor proportion score was reported in every case following the recommendations of the International Association for the Study of Lung Cancer (IASLC). RESULTS In 57 sets of samples, both PD-L1 assays were used. Due to the availability of samples, only 1 assay was performed in 3 sets of samples and in 2 cases, only cytology smears were used and tested for both assays. A total of 113 sets of paired samples finally were evaluated; 4 cases could not be studied due to intense nonspecific background staining. A significant concordance between the 2 assays on cytological smears was found. Concordance between paired cytological smears and formalin-fixed paraffin-embedded samples was observed in 97.3% of the cases. CONCLUSIONS The quantification of PD-L1 expression on direct Papanicolaou-stained cytology smears is feasible and reliable for both PD-L1 assays.
Collapse
Affiliation(s)
- Maria D Lozano
- Department of Pathology, University of Navarra Clinic, University of Navarra, Pamplona, Spain.,CIBERONC, Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain.,Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Marta Abengozar-Muela
- Department of Pathology, University of Navarra Clinic, University of Navarra, Pamplona, Spain
| | - José I Echeveste
- Department of Pathology, University of Navarra Clinic, University of Navarra, Pamplona, Spain
| | - José Carlos Subtil
- Department of Gastroenterology, University of Navarra Clinic, University of Navarra, Pamplona, Spain
| | - Juan Bertó
- Pulmonary Department, University of Navarra Clinic, University of Navarra, Pamplona, Spain
| | - Alfonso Gúrpide
- Department of Medical Oncology, University of Navarra Clinic, University of Navarra, Pamplona, Spain
| | - Alfonso Calvo
- Health Research Institute of Navarra (IDISNA), Pamplona, Spain.,Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Department of Histology and Pathology, University of Navarra, Pamplona, Spain
| | - Carlos E de Andrea
- Department of Pathology, University of Navarra Clinic, University of Navarra, Pamplona, Spain.,CIBERONC, Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain.,Health Research Institute of Navarra (IDISNA), Pamplona, Spain.,Department of Histology and Pathology, University of Navarra, Pamplona, Spain
| |
Collapse
|
15033
|
Characterizing Lymphoma Incidence and Disparities for a Cancer Center Catchment Region. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 19:699-708.e5. [PMID: 31494062 DOI: 10.1016/j.clml.2019.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 06/07/2019] [Accepted: 06/17/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND Racial disparities in non-Hodgkin lymphoma (NHL) are not well-elucidated for specific catchment areas, which can influence outcomes. Leveraging regional data from a population-based cancer registry may provide unique opportunities to quantify NHL disparities. MATERIALS AND METHODS Using Surveillance, Epidemiology, and End Results (SEER) data for NHL cases diagnosed in Georgia from 2001 to 2015, we examined NHL incidence rates by lymphoma subtype and racial differences in baseline characteristics and outcomes for diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL). Cox regression models identified predictors of overall survival (OS). RESULTS SEER documented 38,504 NHL cases in Georgia from 2001 to 2015. The age-adjusted incidence rate for NHL in Georgia increased 1.03% per year, and the annual percentage change was 1.72 in blacks compared with 0.84 in whites. Compared with whites, blacks with DLBCL and FL were more likely to be diagnosed at a younger age (DLBCL, 54.1 vs. 65.5 years; P < .0001; FL, 58.4 vs. 64.0 years; P < .0001) and with B symptoms (DLBCL, 44.4% vs. 33.4%; P < .0001; FL, 28.5% vs. 21.4%; P = .004). Across racial categories, age at diagnosis > 60 years, advanced stage, and B symptoms predicted worse OS in DLBCL and FL. Blacks with DLBCL more commonly were diagnosed with stage III/IV disease (55.5% vs. 48.1%; P < .0001) and had worse 5-year relative survival (58.8% vs. 62.3%; P = .01). CONCLUSIONS Regional cancer registry data can be used to define incidence patterns and disparities in outcomes across NHL subtypes to help define key targets for interventions in a catchment area.
Collapse
|
15034
|
Ailawadhi S, Azzouqa AG, Hodge D, Cochuyt J, Jani P, Ahmed S, Sher T, Roy V, Ailawadhi M, Alegria VR, Manochakian R, Vishnu P, Grover A, Abdulazeez MF, Paulus A, Chanan-Khan A. Survival Trends in Young Patients With Multiple Myeloma: A Focus on Racial-Ethnic Minorities. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 19:619-623. [PMID: 31377212 DOI: 10.1016/j.clml.2019.06.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/05/2019] [Accepted: 06/20/2019] [Indexed: 10/26/2022]
Abstract
INTRODUCTION Outcomes in multiple myeloma (MM) have improved significantly over time. This is true overall for all patients as well as patient subgroups based on age and race/ethnicity. Despite this, disparities are noted in outcomes when looking at racial subgroups. MATERIALS AND METHODS We performed an analysis from the population-based Surveillance, Epidemiology, and End Results (SEER) database to evaluate improvement in relative survival rates (RSRs) for young (≤ 40 years at the time of MM diagnosis) and older (> 40 years at the time of MM diagnosis) over time by race/ethnicity, specifically focusing on Hispanic patients with MM. Expected survival was estimated using the age- and gender-specific death rates from the United States population. RSR was provided as the ratio of the observed to expected survival at individual time points. Five-year and 10-year RSRs were calculated for patients based on treatments modalities available in various time periods. RESULTS We identified a total of 89,451 patients with MM in SEER, of which 1460 patients formed the young patients with MM (≤ 40 years) cohort. Five- and 10-year RSR improved significantly over time for all patients and older patients (> 40 years) by race (all P < .001). Evaluating the younger patients, RSR improved significantly for non-Hispanic whites and non-Hispanic blacks, but not for Hispanics. This was true for the 5-year (P = .08) and 10-year (P = .13) RSRs. CONCLUSION We report a lack of significant benefit in long-term outcomes for younger Hispanic patients with MM over time. This could be owing to multifactorial causes that need to be addressed to mitigate outcome disparities.
Collapse
Affiliation(s)
| | | | - David Hodge
- Department of Health Sciences and Research, Mayo Clinic, Jacksonville, FL
| | - Jordan Cochuyt
- Department of Health Sciences and Research, Mayo Clinic, Jacksonville, FL
| | - Prachi Jani
- Division of Hematology-Oncology, Mayo Clinic, Jacksonville, FL
| | - Salman Ahmed
- Division of Hematology-Oncology, Mayo Clinic, Jacksonville, FL
| | - Taimur Sher
- Division of Hematology-Oncology, Mayo Clinic, Jacksonville, FL
| | - Vivek Roy
- Division of Hematology-Oncology, Mayo Clinic, Jacksonville, FL
| | | | | | | | - Prakash Vishnu
- Division of Hematology-Oncology, Mayo Clinic, Jacksonville, FL
| | - Ashna Grover
- Division of Hematology-Oncology, Mayo Clinic, Jacksonville, FL
| | | | - Aneel Paulus
- Division of Hematology-Oncology, Mayo Clinic, Jacksonville, FL
| | | |
Collapse
|
15035
|
Fan M, Mo T, Shen L, Yang L. Osimertinib-induced severe interstitial lung disease: A case report. Thorac Cancer 2019; 10:1657-1660. [PMID: 31241220 PMCID: PMC6610285 DOI: 10.1111/1759-7714.13127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/03/2019] [Accepted: 06/05/2019] [Indexed: 01/27/2023] Open
Abstract
Lung cancer is globally one of the leading causes of malignant tumor‐related mortality and ranks as having the highest incidence found in men and second in women. Osimertinib is a drug used to target lung cancer but its toxicity is not fully understood. Here we present a case of lung adenocarcinoma in a 78‐year‐old man that was treated and responded favorably with Osimertinib after the failure of other therapies. Unfortunately, the patient developed severe interstitial lung disease during the treatment procedure.
Collapse
Affiliation(s)
- Mengdi Fan
- Department of Endocrinology, Shu Lan (HangZhou) Hospital, Hangzhou, China
| | - Ting Mo
- Department of Respiratory, Shu Lan (HangZhou) Hospital, Hangzhou, China
| | - Lulei Shen
- Department of Respiratory, Shu Lan (HangZhou) Hospital, Hangzhou, China
| | - Li Yang
- Department of Respiratory, Shu Lan (HangZhou) Hospital, Hangzhou, China
| |
Collapse
|
15036
|
Wang H, Ma Y, Lin Y, Liu J, Chen R, Xu B, Liang Y. An Isoxazole Derivative SHU00238 Suppresses Colorectal Cancer Growth through miRNAs Regulation. Molecules 2019; 24:molecules24122335. [PMID: 31242597 PMCID: PMC6630644 DOI: 10.3390/molecules24122335] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 06/22/2019] [Accepted: 06/25/2019] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related deaths worldwide. Isoxazoline and isoxazole derivatives represent an important class of five-membered heterocycles, which play a pivotal role in drug discovery. In our previous study, we developed a series of isoxazole derivatives with an efficient method. In this study, we evaluated their effects on tumor cell growth. HCT116 cells were treated with isoxazole derivatives; an cholecystokinin octapeptide (CCK-8) assay was used to calculate the IC50 (half maximal inhibitory concentration) of each derivative. Compound SHU00238, which was obtained by the copper nitrate-mediated [2+2+1] cycloaddition reaction of olefinic azlactone with naphthalene-1,4-dione, has a lower IC50; we analyzed its inhibitory activity in further assays. Cell apoptosis was estimated by flow cytometry analysis in vitro. SHU00238 injection was used to treat tumor-bearing mice. We found that SHU00238 suppressed cell viability and promoted cell apoptosis in vitro. SHU00238 treatment significantly inhibited colonic tumor growth in vivo. Furthermore, we compared the miRNAs expression changes in HCT116 cells before and after SHU00238 treatment. MiRNA profiling revealed that SHU00238 treatment affected cell fate by regulating a set of miRNAs. In conclusion, SHU00238 impedes CRC tumor cell proliferation and promotes cell apoptosis by miRNAs regulation.
Collapse
Affiliation(s)
- Haoyu Wang
- Department of Chemistry, Qianweichang College, Shanghai University, Shanghai 200444, China.
- School of Life Science, Shanghai University, Shanghai 200444, China.
| | - Yurui Ma
- School of Life Science, Shanghai University, Shanghai 200444, China.
| | - Yifan Lin
- Department of Chemistry, Qianweichang College, Shanghai University, Shanghai 200444, China.
| | - Jiajie Liu
- Department of Chemistry, Qianweichang College, Shanghai University, Shanghai 200444, China.
| | - Rui Chen
- School of Life Science, Shanghai University, Shanghai 200444, China.
| | - Bin Xu
- Department of Chemistry, Qianweichang College, Shanghai University, Shanghai 200444, China.
- Innovative Drug Research Center, Shanghai University, Shanghai 200444, China.
| | - Yajun Liang
- School of Life Science, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
15037
|
Fortunato O, Gasparini P, Boeri M, Sozzi G. Exo-miRNAs as a New Tool for Liquid Biopsy in Lung Cancer. Cancers (Basel) 2019; 11:E888. [PMID: 31242686 PMCID: PMC6627875 DOI: 10.3390/cancers11060888] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/19/2019] [Accepted: 06/24/2019] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is the predominant cause of cancer-related deaths. The high mortality rates are mainly due to the lack of diagnosis before the cancer is at a late stage. Liquid biopsy is a promising technique that could allow early diagnosis of lung cancer and better treatment selection for patients. Cell-free microRNAs have been detected in biological fluids, such as serum and plasma, and are considered interesting biomarkers for lung cancer screening and detection. Exosomes are nanovesicles of 30-150 nm and can be released by different cell types within the tumor microenvironment. Their exosomal composition reflects that of their parental cells and could be potentially useful as a biomarker for lung cancer diagnosis. This review summarizes the state-of-the-art of circulating microRNAs (miRNAs) in lung cancer, focusing on their potential use in clinical practice. Moreover, we describe the importance of exosomal miRNA cargo in lung cancer detection and their potential role during lung carcinogenesis. Finally, we discuss our experience with the analysis of circulating exosomal miRNAs in the bioMILD screening trial.
Collapse
Affiliation(s)
- Orazio Fortunato
- Tumor Genomics Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133 Milan, Italy.
| | - Patrizia Gasparini
- Tumor Genomics Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133 Milan, Italy.
| | - Mattia Boeri
- Tumor Genomics Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133 Milan, Italy.
| | - Gabriella Sozzi
- Tumor Genomics Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133 Milan, Italy.
| |
Collapse
|
15038
|
Zhang Z, Zhu X. Clinical Significance of Lysophosphatidic Acid Receptor-2 (LPA2) and Krüppel-Like Factor 5 (KLF5) Protein Expression Detected by Tissue Microarray in Gastric Adenocarcinoma. Med Sci Monit 2019; 25:4705-4715. [PMID: 31235682 PMCID: PMC6607942 DOI: 10.12659/msm.916336] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background The aim of this study was to evaluate lysophosphatidic acid receptor-2 (LPA2) and Krüppel-like factor 5 (KLF5) protein expression in gastric adenocarcinoma and their correlation with patient clinicopathological characteristics and prognosis. Material/Methods Fifty-one gastric adenocarcinoma tissue samples, 21 gastric intraepithelial neoplasia (GIN) samples, and 13 normal gastric tissue samples were collected to test for LPA2 and KLF5 expression by tissue microarray and immunohistochemistry assay. LPA2 and KLF5 positive expression rate between gastric adenocarcinoma, GIN, and normal gastric tissue were compared. The relationship between LPA2 expression, KLF5 expression, and patients’ clinicopathological characteristics and prognosis were evaluated. Results The positive expression rate of LPA2 and KLF5 were statistical different in gastric adenocarcinoma, GIN, and normal gastric tissue (P<0.05). LPA2 positive expression was associated with tumor invasion depth, Lauren type, vascular invasion, local lymph node metastasis, and clinical stage (P<0.05). There was no correlation between LPA2 expression (hazard ratio [HR]=1.84, 95% confidence interval [CI]: 0.89–3.80, P>0.05), KLF5 expression (HR=1.13, 95% CI: 0.53–2.36, P>0.05), and gastric cancer patients’ overall survival. Conclusions LPA2 and KLF5 protein expressions were differently expressed in gastric adenocarcinoma, GIN, and normal gastric tissue, and differences were correlated with patients’ clinical characteristic. However, LPA2 and KLF5 expressions were not correlated with the patients’ prognosis.
Collapse
Affiliation(s)
- Zhili Zhang
- Department of Pathology, The Second People's Hospital of Jiuquan, Jiuquan, Gansu, China (mainland)
| | - Xiaoyong Zhu
- Departments of Oncology, Zhuji People's Hospital of Zhejiang Province, Zhuji, Zhejiang, China (mainland)
| |
Collapse
|
15039
|
Yu Q, Wang P, Yang L, Wu Z, Li S, Xu Y, Wu B, Ma A, Gan X, Xu R. Novel synthetic tosyl chloride-berbamine regresses lethal MYC-positive leukemia by targeting CaMKIIγ/Myc axis. Biomed Pharmacother 2019; 117:109134. [PMID: 31247466 DOI: 10.1016/j.biopha.2019.109134] [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] [Received: 04/29/2019] [Revised: 06/13/2019] [Accepted: 06/13/2019] [Indexed: 01/05/2023] Open
Abstract
Proto-oncogene Myc, a key transcription factor, is frequently deregulated in human leukemia with aggressive and poor clinical outcome, but the development of MYC inhibitors remains challenging due to MYC helix-loop-helix topology lacking druggable domains. Here we describe a novel oral active small molecule analog of berbamine, tosyl chloride-berbamine (TCB), that efficiently eliminates MYC-positive leukemia in vitro and in vivo. Mechanistically, TCB potently reduced MYC protein by inhibiting CaMKIIγ, a critical enzyme that stabilizes MYC protein, and induces apoptosis of MYC-positive leukemia cells. In vivo, oral administration of TCB markedly eliminated lethal MYC-positive acute lymphoblastic leukemia (ALL) with well tolerability in orthotopic mouse model. Our studies identify CaMKIIγ/Myc axis as a valid target for developing small molecule-based new therapies for treating MYC-mediated leukemia and demonstrate that TCB is an orally active analog of berbamine that kills MYC-positive leukemia cells.
Collapse
Affiliation(s)
- Qingfeng Yu
- Department of Hematology and Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Ping Wang
- Department of Hematology and Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Linlin Yang
- Department of Hematology and Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Zhaoxing Wu
- Department of Hematology and Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Shu Li
- Department of Hematology and Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Ying Xu
- Department of Hematology and Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Bowen Wu
- Department of Hematology and Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - An Ma
- Zhejiang Academy of Medical Sciences, Hangzhou, 310012, China
| | - Xiaoxian Gan
- Zhejiang Academy of Medical Sciences, Hangzhou, 310012, China
| | - Rongzhen Xu
- Department of Hematology and Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China; Institute of Hematology, Zhejiang University, Hangzhou, 310009, China.
| |
Collapse
|
15040
|
Marinac CR, Suppan CA, Giovannucci E, Song M, Kværner AS, Townsend MK, Rosner BA, Rebbeck TR, Colditz GA, Birmann BM. Elucidating Under-Studied Aspects of the Link Between Obesity and Multiple Myeloma: Weight Pattern, Body Shape Trajectory, and Body Fat Distribution. JNCI Cancer Spectr 2019; 3:pkz044. [PMID: 31448358 PMCID: PMC6699596 DOI: 10.1093/jncics/pkz044] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 05/30/2019] [Accepted: 06/19/2019] [Indexed: 11/14/2022] Open
Abstract
Background Although obesity is an established modifiable risk factor for multiple myeloma (MM), several nuanced aspects of its relation to MM remain unelucidated, limiting public health and prevention messages. Methods We analyzed prospective data from the Nurses' Health Study and Health Professionals Follow-Up Study to examine MM risk associated with 20-year weight patterns in adulthood, body shape trajectory from ages 5 to 60 years, and body fat distribution. For each aforementioned risk factor, we report hazard ratios (HRs) and 95% confidence intervals (CIs) for incident MM from multivariable Cox proportional-hazards models. Results We documented 582 incident MM cases during 4 280 712 person-years of follow-up. Persons who exhibited extreme weight cycling, for example, those with net weight gain and one or more episodes of intentional loss of at least 20 pounds or whose cumulative intentional weight loss exceeded net weight loss with at least one episode of intentional loss of 20 pounds or more had an increased MM risk compared with individuals who maintained their weight (HR = 1.71, 95% CI = 1.05 to 2.80); the association was statistically nonsignificant after adjustment for body mass index. We identified four body shape trajectories: lean-stable, lean-increase, medium-stable, and medium-increase. MM risk was higher in the medium-increase group than in the lean-stable group (HR = 1.62, 95% CI = 1.22 to 2.14). Additionally, MM risk increased with increasing hip circumference (HR per 1-inch increase: 1.03, 95% CI = 1.01 to 1.06) but was not associated with other body fat distribution measures. Conclusions Maintaining a lean and stable weight throughout life may provide the strongest benefit in terms of MM prevention.
Collapse
Affiliation(s)
| | | | | | - Mingyang Song
- See the Notes section for the full list of authors' affiliations
| | - Ane S Kværner
- See the Notes section for the full list of authors' affiliations
| | - Mary K Townsend
- See the Notes section for the full list of authors' affiliations
| | - Bernard A Rosner
- See the Notes section for the full list of authors' affiliations
| | | | - Graham A Colditz
- See the Notes section for the full list of authors' affiliations
| | - Brenda M Birmann
- See the Notes section for the full list of authors' affiliations
| |
Collapse
|
15041
|
Zhao H, Zhao C, Li H, Zhang D, Liu G. E2A attenuates tumor-initiating capacity of colorectal cancer cells via the Wnt/beta-catenin pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:276. [PMID: 31234887 PMCID: PMC6591938 DOI: 10.1186/s13046-019-1261-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/31/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND The E2A gene, which encodes two basic helix-loop-helix transcription factors, E12 and E47, regulates colorectal cancer progression and epithelial-mesenchymal transition. However, whether E2A regulates the tumor-initiating capacity of colorectal cancer is unclear. Thus, we have studied E2A expression in the initiation of colorectal cancer in vivo and in vitro. METHODS Immunohistochemistry and immunoblot were performed to determine protein levels of E2A in colorectal cancer specimens and cells. RNAi was employed to downregulate E2A expression, and the subsequent change in protein level was evaluated by immunoblot. Sphere-forming assay and enumeration of liver metastasis in mouse models were used to identify the tumor formation ability of colorectal cancer cells. RESULTS E2A expression in colorectal cancer clinical specimens was inversely associated with patients' progression-free survival. Functional studies demonstrated that E2A significantly decreased tumor formation in vitro and in vivo. Furthermore, nuclear translocation of beta-catenin and activation of the Wnt/beta-catenin pathway occurred after suppression of E2A in colorectal cancer cells. FoxM1 was identified as a down-stream target by mRNA microarray, implying that FoxM1 plays a main role in determining how E2A regulates the tumor-initiating capacity of colorectal cancer. CONCLUSION E2A suppresses tumor-initiating capacity by targeting the FoxM1-Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Hongchao Zhao
- Department of General Surgery, The First Affiliated Hospital of Zhengzhou University, 41 Jianshe Road, Zhengzhou, Henan, China
| | - Chunlin Zhao
- Department of General Surgery, The First Affiliated Hospital of Zhengzhou University, 41 Jianshe Road, Zhengzhou, Henan, China
| | - Haohao Li
- Department of General Surgery, The First Affiliated Hospital of Zhengzhou University, 41 Jianshe Road, Zhengzhou, Henan, China
| | - Danhua Zhang
- Department of General Surgery, The First Affiliated Hospital of Zhengzhou University, 41 Jianshe Road, Zhengzhou, Henan, China.
| | - Guanghui Liu
- Department of General Surgery, The First Affiliated Hospital of Zhengzhou University, 41 Jianshe Road, Zhengzhou, Henan, China.
| |
Collapse
|
15042
|
Lee JW, Beatty GL. Hepatocytes prepare the soil for liver metastasis. Mol Cell Oncol 2019; 6:e1632686. [PMID: 31528701 DOI: 10.1080/23723556.2019.1632686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 01/25/2023]
Abstract
The liver is the most common organ site of cancer metastasis. Molecular determinants of this organotropism, though, are poorly understood. We recently showed that hepatocytes regulate the formation of a "pro-metastatic" niche in the liver via their release of serum amyloid proteins.
Collapse
Affiliation(s)
- Jae W Lee
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gregory L Beatty
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
15043
|
Wang G, Zhao J, Zhang M, Wang Q, Chen B, Hou Y, Lu K. Ferumoxytol and CpG oligodeoxynucleotide 2395 synergistically enhance antitumor activity of macrophages against NSCLC with EGFR L858R/T790M mutation. Int J Nanomedicine 2019; 14:4503-4515. [PMID: 31417255 PMCID: PMC6599896 DOI: 10.2147/ijn.s193583] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 04/22/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose: Drug resistance is a major challenge for epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) treatment of lung cancer. Ferumoxytol (FMT) drives macrophage (MΦ) transformation towards a M1-like phenotype and thereby inhibits tumor growth. CpG oligodeoxynucleotide 2395 (CpG), a toll-like receptor 9 (TLR9) agonist, is an effective therapeutic agent to induce anticancer immune responses. Herein, the effect of co-administered FMT and CpG on MΦ activation for treating non-small cell lung cancer (NSCLC) was explored. Methods: The mRNA expression levels of M1-like genes in RAW 264.7 MΦ cells stimulated by FMT, CpG and FMT and CpG (FMT/CpG) were evaluated by quantitative reverse transcription PCR (qRT-PCR). Then, the effects of FMT/CpG-pretreated MΦ supernatant on apoptosis and proliferation of H1975 cells were detected by flow cytometry, and the expression of EGFR and its downstream signaling pathway in H1975 cells were explored by western blotting. Finally, a H1975 cell xenograft mouse model was used to study the anti-tumor effect of the combination of FMT and CpG in vivo. Results: FMT and CpG synergistically enhanced M1-like gene expression in MΦ, including tumor necrosis factor-α, interleukin (IL)-12, IL-1α, IL-1β, IL-6 and inducible nitric oxide synthase (iNOS). FMT/CpG-pretreated MΦ supernatant inhibited proliferation and induced apoptosis of H1975 cells, accompanied by down-regulation of cell cycle-associated proteins and up-regulation of apoptosis-related proteins. Further studies indicated that the FMT/CpG-pretreated MΦ supernatant suppressed p-EGFR and its downstream AKT/mammalian target of rapamycin signaling pathway in H1975 cells. Furthermore, FMT/CpG suppressed tumor growth in mice accompanied by a decline in the EGFR-positive tumor cell fraction and increased M1 phenotype macrophage infiltration. Conclusion: FMT acted synergistically with CpG to activate MΦ for suppressed proliferation and promoted apoptosis of NSCLC cells via EGFR signaling. Thus, combining FMT and CpG is an effective strategy for the treatment of NSCLC with EGFRL858R/T790M mutation.
Collapse
Affiliation(s)
- Guoqun Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Jiaojiao Zhao
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, People's Republic of China
| | - Meiling Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Qian Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Bo Chen
- Institute of Materials Science and Devices, Suzhou University of Science and Technology, Suzhou 215009, People's Republic of China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, People's Republic of China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, People's Republic of China
| | - Kaihua Lu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| |
Collapse
|
15044
|
Hung YH, Hsu MC, Chen LT, Hung WC, Pan MR. Alteration of Epigenetic Modifiers in Pancreatic Cancer and Its Clinical Implication. J Clin Med 2019; 8:jcm8060903. [PMID: 31238554 PMCID: PMC6617267 DOI: 10.3390/jcm8060903] [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: 05/30/2019] [Revised: 06/15/2019] [Accepted: 06/20/2019] [Indexed: 12/12/2022] Open
Abstract
The incidence of pancreatic cancer has considerably increased in the past decade. Pancreatic cancer has the worst prognosis among the cancers of the digestive tract because the pancreas is located in the posterior abdominal cavity, and most patients do not show clinical symptoms for early detection. Approximately 55% of all patients are diagnosed with pancreatic cancer only after the tumors metastasize. Therefore, identifying useful biomarkers for early diagnosis and screening high-risk groups are important to improve pancreatic cancer therapy. Recent emerging evidence has suggested that genetic and epigenetic alterations play a crucial role in the molecular aspects of pancreatic tumorigenesis. Here, we summarize recent progress in our understanding of the epigenetic alterations in pancreatic cancer and propose potential synthetic lethal strategies to target these genetic defects to treat this deadly disease.
Collapse
Affiliation(s)
- Yu-Hsuan Hung
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| | - Ming-Chuan Hsu
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
- Division of Hematology/Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan 704, Taiwan.
| | - Wen-Chun Hung
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
- Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Mei-Ren Pan
- Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| |
Collapse
|
15045
|
Yang F, Shao C, Wei K, Jing X, Qin Z, Shi Y, Shu Y, Shen H. miR-942 promotes tumor migration, invasion, and angiogenesis by regulating EMT via BARX2 in non-small-cell lung cancer. J Cell Physiol 2019; 234:23596-23607. [PMID: 31236953 DOI: 10.1002/jcp.28928] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/21/2019] [Accepted: 05/22/2019] [Indexed: 12/14/2022]
Abstract
Epithelial-mesenchymal transition (EMT) has an important function in cancer. Recently, microRNAs have been reported to be involved in EMT by regulating target genes. miR-942 is considered a novel oncogene in esophageal squamous cell carcinoma. However, its role in non-small-cell lung cancer (NSCLC) has not been investigated. In this study, the expression of miR-942 in NSCLC patients tumor and paired adjacent tissues were assessed by quantitative real-time polymerase chain reaction and in situ hybridization. Transwell, wound healing, tube formation, and tail vein xenograft assays were conducted to assess miR-942's function in NSCLC. Potential miR-942 targets were confirmed using dual-luciferase reporter assays, immunohistochemistry, immunoblot, and rescue experiments. The results showed miR-942 is relatively highly expressed in human NSCLC tissues and cells. In vitro assays demonstrated that overexpression of miR-942 promoted cell migration, invasion, and angiogenesis. Tail vein xenograft assays suggested that miR-942 contributed to NSCLC metastasis in vivo. Three bioinformatics software was searched, and BARX2 was predicted as a downstream target of miR-942. Direct interaction between them was validated by dual-luciferase assays. Rescue experiments further confirmed that BARX2 overexpression could reverse functional changes caused by miR-942. Moreover, miR-942 increased EMT-associated proteins N-cadherin and vimentin by inhibiting BARX2, while E-cadherin expression is reduced. In summary, this study reveals that miR-942 induces EMT-related metastasis by directly targeting BARX2, which may provide a potential therapeutic strategy for NSCLC.
Collapse
Affiliation(s)
- Fengming Yang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Oncology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Chuchu Shao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Oncology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Ke Wei
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinming Jing
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhiqiang Qin
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yuenian Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Oncology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Hua Shen
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Oncology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
15046
|
Wondergem M, Jansen BHE, van der Zant FM, van der Sluis TM, Knol RJJ, van Kalmthout LWM, Hoekstra OS, van Moorselaar RJA, Oprea-Lager DE, Vis AN. Early lesion detection with 18F-DCFPyL PET/CT in 248 patients with biochemically recurrent prostate cancer. Eur J Nucl Med Mol Imaging 2019; 46:1911-1918. [PMID: 31230088 PMCID: PMC6647179 DOI: 10.1007/s00259-019-04385-6] [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: 03/14/2019] [Accepted: 05/30/2019] [Indexed: 01/14/2023]
Abstract
Purpose Prostate-specific membrane antigen (PSMA) PET/CT is increasingly used in patients with biochemically recurrent prostate cancer (BCR), mostly using gallium-68 (168Ga)-labelled radiotracers. Alternatively, fluorine-18 (18F)-labelled PSMA tracers are available, such as 18F-DCFPyL, which offer enhanced image quality and therefore potentially increased detection of small metastases. In this study we evaluate the lesion detection efficacy of 18F-DCFPyL PET/CT in patients with BCR and determine the detection efficacy as a function of their PSA value. Methods A total of 248 consecutive patients were evaluated and underwent scanning with 18F-DCFPyL PET/CT for BCR between November 2016 and 2018 in two hospitals in the Netherlands. Patients were examined after radical prostatectomy (52%), external-beam radiation therapy (42%) or brachytherapy (6%). Imaging was performed 120 min after injection of a median dose of 311 MBq 18F-DCFPyL. Results In 214 out of 248 PET/CT scans (86.3%), at least one lesion suggestive of cancer recurrence was detected (‘positive scan’). Scan positivity increased with higher PSA values: 17/29 scans (59%) with PSA values <0.5 ng/ml; 20/29 (69%) with PSA 0.5 to <1.0 ng/ml; 35/41 (85%) with PSA 1.0 to <2.0 ng/ml; 69/73 (95%) with PSA 2.0 to <5.0 ng/ml; and 73/76 (96%) with PSA ≥5.0 ng/ml. Interestingly, suspicious lesions outside the prostatic fossa were detected in 39–50% of patients with PSA <1.0 ng/ml after radical prostatectomy (i.e. candidates for salvage radiotherapy). Conclusion 18F-DCFPyL PET/CT offers early detection of lesions in patients with BCR, even at PSA levels <0.5 ng/ml. These results appear to be comparable to those reported for 68Ga-PSMA and 18F-PSMA-1007, with potentially increased detection efficacy compared to 68Ga-PSMA for patients with PSA <2.0. Electronic supplementary material The online version of this article (10.1007/s00259-019-04385-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- M Wondergem
- Noordwest Ziekenhuisgroep, Nuclear Medicine, Wilhelminalaan 12, 1815 JD, Alkmaar, the Netherlands
| | - B H E Jansen
- Amsterdam University Medical Centers, VU University, Urology, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands. .,Amsterdam University Medical Centers, VU University, Radiology & Nuclear Medicine, Amsterdam, the Netherlands.
| | - F M van der Zant
- Noordwest Ziekenhuisgroep, Nuclear Medicine, Wilhelminalaan 12, 1815 JD, Alkmaar, the Netherlands
| | - T M van der Sluis
- Amsterdam University Medical Centers, VU University, Urology, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| | - R J J Knol
- Noordwest Ziekenhuisgroep, Nuclear Medicine, Wilhelminalaan 12, 1815 JD, Alkmaar, the Netherlands
| | - L W M van Kalmthout
- Radiology & Nuclear Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
| | - O S Hoekstra
- Amsterdam University Medical Centers, VU University, Radiology & Nuclear Medicine, Amsterdam, the Netherlands
| | - R J A van Moorselaar
- Amsterdam University Medical Centers, VU University, Urology, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| | - D E Oprea-Lager
- Amsterdam University Medical Centers, VU University, Radiology & Nuclear Medicine, Amsterdam, the Netherlands
| | - A N Vis
- Amsterdam University Medical Centers, VU University, Urology, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| |
Collapse
|
15047
|
Ghandour R, Singla N, Lotan Y. Treatment Options and Outcomes in Nonmetastatic Muscle Invasive Bladder Cancer. Trends Cancer 2019; 5:426-439. [PMID: 31311657 DOI: 10.1016/j.trecan.2019.05.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/23/2019] [Accepted: 05/30/2019] [Indexed: 12/26/2022]
Abstract
Muscle-invasive bladder cancer (MIBC) represents 25% of newly diagnosed bladder cancer. MIBC is aggressive and requires timely management. The current standard of care is neoadjuvant chemotherapy followed by radical cystectomy, an approach that could result in significant morbidities. Modifications in the chemotherapy regimens, as well as in perioperative care and surgical approach, have resulted in better overall toxicity profile and faster recovery. However, bladder-preservation in carefully selected patients can lead to acceptable oncological outcomes and better quality of life. Optimization of bladder-preservation protocols and proper identification of patients who tolerate and respond to various treatment modalities will significantly impact patient survival in the coming future.
Collapse
Affiliation(s)
- Rashed Ghandour
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9110, USA
| | - Nirmish Singla
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9110, USA
| | - Yair Lotan
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9110, USA.
| |
Collapse
|
15048
|
PDS5B regulates cell proliferation and motility via upregulation of Ptch2 in pancreatic cancer cells. Cancer Lett 2019; 460:65-74. [PMID: 31233836 DOI: 10.1016/j.canlet.2019.06.014] [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: 03/18/2019] [Revised: 06/18/2019] [Accepted: 06/19/2019] [Indexed: 12/19/2022]
Abstract
Pds5b (precocious dissociation of sisters 5B) is involved in both tumorigenesis and cancer progression; however, the functions and molecular mechanisms of Pds5b in pancreatic cancer (PC) are unknown. Several approaches were conducted to investigate the molecular basis of Pds5b-related PC progression, including transfection, MTT, FACS, western blotting, wound healing assay, transwell chamber invasion assay, and immunohistochemical methods. Pds5b overexpression inhibited cell growth and induced apoptosis, whereas the inhibition of Pds5b promoted growth of PC cells. Moreover, Pds5b overexpression inhibited cell migration and invasion, while the downregulation of Pds5b enhanced cell motility. Furthermore, reduced Pds5b expression was associated with survival in PC patients. Mechanistically, Pds5b positively regulated the expression of Ptch2 to influence the Sonic hedgehog signaling pathway. Consistently, Ptch2 downregulation enhanced cell growth, migration, and invasion, while inhibiting cell apoptosis. Notably, the downregulation of Ptch2 abolished Pds5b-mediated anti-tumor activity in PC cells. Strikingly, Pds5b expression was positively associated with levels of Ptch2 in PC patient samples, suggesting that the Pds5b/Ptch2 axis regulates cell proliferation and invasion in PC cells. Our findings indicate that targeting Pds5b and Ptch2 may represent a novel therapeutic approach for PC.
Collapse
|
15049
|
Warner JL, Dymshyts D, Reich CG, Gurley MJ, Hochheiser H, Moldwin ZH, Belenkaya R, Williams AE, Yang PC. HemOnc: A new standard vocabulary for chemotherapy regimen representation in the OMOP common data model. J Biomed Inform 2019; 96:103239. [PMID: 31238109 DOI: 10.1016/j.jbi.2019.103239] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/20/2019] [Accepted: 06/21/2019] [Indexed: 10/26/2022]
Abstract
Systematic application of observational data to the understanding of impacts of cancer treatments requires detailed information models allowing meaningful comparisons between treatment regimens. Unfortunately, details of systemic therapies are scarce in registries and data warehouses, primarily due to the complex nature of the protocols and a lack of standardization. Since 2011, we have been creating a curated and semi-structured website of chemotherapy regimens, HemOnc.org. In coordination with the Observational Health Data Sciences and Informatics (OHDSI) Oncology Subgroup, we have transformed a substantial subset of this content into the OMOP common data model, with bindings to multiple external vocabularies, e.g., RxNorm and the National Cancer Institute Thesaurus. Currently, there are >73,000 concepts and >177,000 relationships in the full vocabulary. Content related to the definition and composition of chemotherapy regimens has been released within the ATHENA tool (athena.ohdsi.org) for widespread utilization by the OHDSI membership. Here, we describe the rationale, data model, and initial contents of the HemOnc vocabulary along with several use cases for which it may be valuable.
Collapse
Affiliation(s)
- Jeremy L Warner
- Vanderbilt University Medical Center, Nashville, TN, United States; HemOnc.org, LLC, Lexington, MA, United States.
| | | | | | | | | | - Zachary H Moldwin
- University of Illinois at Chicago College of Pharmacy, Chicago, IL, United States
| | - Rimma Belenkaya
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | - Peter C Yang
- HemOnc.org, LLC, Lexington, MA, United States; Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
15050
|
Multidisciplinary Management of Patients with Unresectable Hepatocellular Carcinoma: A Critical Appraisal of Current Evidence. Cancers (Basel) 2019; 11:cancers11060873. [PMID: 31234476 PMCID: PMC6627394 DOI: 10.3390/cancers11060873] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/19/2019] [Accepted: 06/21/2019] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of new cancer diagnoses in the United States, with an incidence that is expected to rise. The etiology of HCC is varied and can lead to differences between patients in terms of presentation and natural history. Subsequently, physicians treating these patients need to consider a variety of disease and patient characteristics when they select from the many different treatment options that are available for these patients. At the same time, the treatment landscape for patients with HCC, particularly those with unresectable HCC, has been rapidly evolving as new, evidence-based options become available. The treatment plan for patients with HCC can include surgery, transplant, ablation, transarterial chemoembolization, transarterial radioembolization, radiation therapy, and/or systemic therapies. Implementing these different modalities, where the optimal sequence and/or combination has not been defined, requires coordination between physicians with different specialties, including interventional radiologists, hepatologists, and surgical and medical oncologists. As such, the implementation of a multidisciplinary team is necessary to develop a comprehensive care plan for patients, especially those with unresectable HCC.
Collapse
|