1
|
Ramu A, Ak L, Chinnappan J. Identification of prostate cancer associated genes for diagnosis and prognosis: a modernized in silico approach. Mamm Genome 2024; 35:683-710. [PMID: 39153107 DOI: 10.1007/s00335-024-10060-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/06/2024] [Indexed: 08/19/2024]
Abstract
Prostate cancer (PCa) ranks as the second leading cause of cancer-related deaths in men. Diagnosing PCa relies on molecular markers known as diagnostic biomarkers, while prognostic biomarkers are used to identify key proteins involved in PCa treatments. This study aims to gather PCa-associated genes and assess their potential as either diagnostic or prognostic biomarkers for PCa. A corpus of 152,064 PCa-related data from PubMed, spanning from May 1936 to December 2020, was compiled. Additionally, 4199 genes associated with PCa terms were collected from the National Center of Biotechnology Information (NCBI) database. The PubMed corpus data was extracted using pubmed.mineR to identify PCa-associated genes. Network and pathway analyses were conducted using various tools, such as STRING, DAVID, KEGG, MCODE 2.0, cytoHubba app, CluePedia, and ClueGO app. Significant marker genes were identified using Random Forest, Support Vector Machines, Neural Network algorithms, and the Cox Proportional Hazard model. This study reports 3062 unique PCa-associated genes along with 2518 corresponding unique PMIDs. Diagnostic markers such as IL6, MAPK3, JUN, FOS, ACTB, MYC, and TGFB1 were identified, while prognostic markers like ACTB and HDAC1 were highlighted in PubMed. This suggests that the potential target genes provided by PubMed data outweigh those in the NCBI database.
Collapse
Affiliation(s)
- Akilandeswari Ramu
- Anthropology and Health Informatics Laboratory, Department of Bioinformatics, Bharathiar University, Coimbatore, Tamil Nadu, India.
| | - Lekhashree Ak
- Anthropology and Health Informatics Laboratory, Department of Bioinformatics, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Jayaprakash Chinnappan
- Anthropology and Health Informatics Laboratory, Department of Bioinformatics, Bharathiar University, Coimbatore, Tamil Nadu, India
| |
Collapse
|
2
|
Savvidis C, Kallistrou E, Kouroglou E, Dionysopoulou S, Gavriiloglou G, Ragia D, Tsiama V, Proikaki S, Belis K, Ilias I. Circadian rhythm disruption and endocrine-related tumors. World J Clin Oncol 2024; 15:818-834. [PMID: 39071458 PMCID: PMC11271730 DOI: 10.5306/wjco.v15.i7.818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
This review delved into the intricate relationship between circadian clocks and physiological processes, emphasizing their critical role in maintaining homeostasis. Orchestrated by interlocked clock genes, the circadian timekeeping system regulates fundamental processes like the sleep-wake cycle, energy metabolism, immune function, and cell proliferation. The central oscillator in the hypothalamic suprachiasmatic nucleus synchronizes with light-dark cycles, while peripheral tissue clocks are influenced by cues such as feeding times. Circadian disruption, linked to modern lifestyle factors like night shift work, correlates with adverse health outcomes, including metabolic syndrome, cardiovascular diseases, infections, and cancer. We explored the molecular mechanisms of circadian clock genes and their impact on metabolic disorders and cancer pathogenesis. Specific associations between circadian disruption and endocrine tumors, spanning breast, ovarian, testicular, prostate, thyroid, pituitary, and adrenal gland cancers, are highlighted. Shift work is associated with increased breast cancer risk, with PER genes influencing tumor progression and drug resistance. CLOCK gene expression correlates with cisplatin resistance in ovarian cancer, while factors like aging and intermittent fasting affect prostate cancer. Our review underscored the intricate interplay between circadian rhythms and cancer, involving the regulation of the cell cycle, DNA repair, metabolism, immune function, and the tumor microenvironment. We advocated for integrating biological timing into clinical considerations for personalized healthcare, proposing that understanding these connections could lead to novel therapeutic approaches. Evidence supports circadian rhythm-focused therapies, particularly chronotherapy, for treating endocrine tumors. Our review called for further research to uncover detailed connections between circadian clocks and cancer, providing essential insights for targeted treatments. We emphasized the importance of public health interventions to mitigate lifestyle-related circadian disruptions and underscored the critical role of circadian rhythms in disease mechanisms and therapeutic interventions.
Collapse
Affiliation(s)
- Christos Savvidis
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Efthymia Kallistrou
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Eleni Kouroglou
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Sofia Dionysopoulou
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | | | - Dimitra Ragia
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Vasiliki Tsiama
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Stella Proikaki
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Konstantinos Belis
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Ioannis Ilias
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| |
Collapse
|
3
|
Hu Y, Fan S, Zhu Y, Xie X. A novel circadian cycle-related gene signature for prognosis prediction of patients with breast cancer. Medicine (Baltimore) 2023; 102:e33718. [PMID: 37144994 PMCID: PMC10158864 DOI: 10.1097/md.0000000000033718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/18/2023] [Indexed: 05/06/2023] Open
Abstract
The extensive and intricate relationships between circadian rhythm and cancer have been reported in numerous studies. However, in breast cancer (BC), the potential role of circadian clock-related genes (CCRGs) in prognosis prediction has not been fully clarified. The transcriptome data and clinical information were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus databases. A CCRGs-based risk signature was established by differential expression analysis, univariate, Lasso and multivariate Cox regression analyses. we conducted a gene set enrichment analysis (GSEA) between groups. A nomogram integrating independent clinical factors and risk score was generated and evaluated by calibration curves and decision curve analysis (DCA). Differentially expression analysis revealed 80 differentially expressed CCRGs, and 27 of them were significantly associated with the overall survival (OS) of BC. BC can be classified into 4 molecular subtypes with significant differences in prognosis based on the 27 CCRGs. Three prognostic CCRGs, including desmocollin 1 (DSC1), LEF1, and protocadherin 9 (PCDH9), were identified to be independent risk factors of BC prognosis and were used to construct a risk score model. BC patients were divided into high- and low-risk groups, and there were significant differences in prognosis between the 2 groups both in the training and validation cohorts. It was found that patients in different groups of race, status, or T stage had significant levels of risk score. Furthermore, patients of different risk levels exhibit varying degrees of sensitivity to vinorelbine, lapatinib, metformin, and vinblastine. GSEA showed that in the high-risk group, immune response-related activities were dramatically repressed whereas cilium-related processes were significantly stimulated. Cox regression analysis demonstrated that age, N stage, radiotherapy and the risk score were independent prognostic risk factors of BC, and a nomogram was established based on these variables. The nomogram exerted a favorable concordance index (0.798) as well as calibration performance, which strongly supports the clinical application of the nomogram. Our study indicated the disruption of the expression of CCRGs in BC and built a favorable prognostic risk model based on 3 independent prognostic CCRGs. These genes may be applied as candidate molecular targets for the diagnosis and therapy of BC.
Collapse
Affiliation(s)
- Yuanyuan Hu
- Department of Breast Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Shuyao Fan
- Department of Breast Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Yiwan Zhu
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiaohong Xie
- Department of Breast Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Zhu WZ, He QY, Feng DC, Wei Q, Yang L. Circadian rhythm in prostate cancer: time to take notice of the clock. Asian J Androl 2023; 25:184-191. [PMID: 36073562 PMCID: PMC10069698 DOI: 10.4103/aja202255] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The circadian clock is an evolutionary molecular product that is associated with better adaptation to changes in the external environment. Disruption of the circadian rhythm plays a critical role in tumorigenesis of many kinds of cancers, including prostate cancer (PCa). Integrating circadian rhythm into PCa research not only brings a closer understanding of the mechanisms of PCa but also provides new and effective options for the precise treatment of patients with PCa. This review begins with patterns of the circadian clock, highlights the role of the disruption of circadian rhythms in PCa at the epidemiological and molecular levels, and discusses possible new approaches to PCa therapy that target the circadian clock.
Collapse
Affiliation(s)
- Wei-Zhen Zhu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qi-Ying He
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - De-Chao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lu Yang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
5
|
Briggs LG, Steele GL, Qian ZJ, Subbana S, Alkhatib KY, Labban M, Langbein BJ, Nguyen DD, Cellini J, Kilbridge K, Kibel AS, Trinh QD, Rana HQ, Cole AP. Racial Differences in Germline Genetic Testing for Prostate Cancer: A Systematic Review. JCO Oncol Pract 2023; 19:e784-e793. [PMID: 36649495 DOI: 10.1200/op.22.00634] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
PURPOSE Testing for pathogenic variants can aid in oncologic risk stratification and identification of targeted therapies. Despite known disparities in access to prostate cancer (PCa) care, little has been written about access to germline genetic testing (GGT) for Black men and other historically marginalized populations. This systematic review sought to delineate racial/ethnic disparities in GGT for PCa. METHODS This systematic review identified articles published from January 1996 through May 2021 in PubMed, Web of Science, and Embase. We included studies that reported rates of GGT in men with PCa in the United States by race/ethnicity as reflective of routine clinical care or research. A narrative synthesis was performed. RESULTS Of 4,309 unique records, 91 studies examining 50 unique study populations met inclusion criteria. Of these, four populations included men who received GGT through routine clinical care, accounting for 4,415 men (72.6% White and 7.2% Black). The other 46 populations included men who received GGT as part of a research study, accounting for 30,824 men (64.3% White and 21.6% Black). Of these 46 research populations, 19 used targeted methods to increase recruitment from a specific demographic. CONCLUSION Most studies that report GGT rates by race/ethnicity are in research settings. Many of these studies used targeted recruitment methods and subsequently have a greater proportion of Black men than clinical and US population-based studies. Other historically marginalized populations are not well represented. There remains a knowledge gap regarding the extent of racial disparities in the use of GGT, particularly in the clinical setting.
Collapse
Affiliation(s)
- Logan G Briggs
- Department of Urologic Surgery, Mayo Clinic, Phoenix, AZ.,Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Grant L Steele
- Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Zhiyu Jason Qian
- Division of Urological Surgery, Brigham and Women's Hospital, Boston, MA
| | | | - Khalid Y Alkhatib
- Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Muhieddine Labban
- Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Bjoern J Langbein
- Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - David-Dan Nguyen
- Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | - Kerry Kilbridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Adam S Kibel
- Division of Urological Surgery, Brigham and Women's Hospital, Boston, MA
| | - Quoc-Dien Trinh
- Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Division of Urological Surgery, Brigham and Women's Hospital, Boston, MA
| | - Huma Q Rana
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.,Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA
| | - Alexander P Cole
- Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Division of Urological Surgery, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
6
|
Jiang Y, Meyers TJ, Emeka AA, Cooley LF, Cooper PR, Lancki N, Helenowski I, Kachuri L, Lin DW, Stanford JL, Newcomb LF, Kolb S, Finelli A, Fleshner NE, Komisarenko M, Eastham JA, Ehdaie B, Benfante N, Logothetis CJ, Gregg JR, Perez CA, Garza S, Kim J, Marks LS, Delfin M, Barsa D, Vesprini D, Klotz LH, Loblaw A, Mamedov A, Goldenberg SL, Higano CS, Spillane M, Wu E, Carter HB, Pavlovich CP, Mamawala M, Landis T, Carroll PR, Chan JM, Cooperberg MR, Cowan JE, Morgan TM, Siddiqui J, Martin R, Klein EA, Brittain K, Gotwald P, Barocas DA, Dallmer JR, Gordetsky JB, Steele P, Kundu SD, Stockdale J, Roobol MJ, Venderbos LD, Sanda MG, Arnold R, Patil D, Evans CP, Dall’Era MA, Vij A, Costello AJ, Chow K, Corcoran NM, Rais-Bahrami S, Phares C, Scherr DS, Flynn T, Karnes RJ, Koch M, Dhondt CR, Nelson JB, McBride D, Cookson MS, Stratton KL, Farriester S, Hemken E, Stadler WM, Pera T, Banionyte D, Bianco FJ, Lopez IH, Loeb S, Taneja SS, Byrne N, Amling CL, Martinez A, Boileau L, Gaylis FD, Petkewicz J, Kirwen N, Helfand BT, Xu J, Scholtens DM, Catalona WJ, Witte JS. Genetic Factors Associated with Prostate Cancer Conversion from Active Surveillance to Treatment. HGG ADVANCES 2022; 3:100070. [PMID: 34993496 PMCID: PMC8725988 DOI: 10.1016/j.xhgg.2021.100070] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/12/2021] [Indexed: 12/18/2022] Open
Abstract
Men diagnosed with low-risk prostate cancer (PC) are increasingly electing active surveillance (AS) as their initial management strategy. While this may reduce the side effects of treatment for prostate cancer, many men on AS eventually convert to active treatment. PC is one of the most heritable cancers, and genetic factors that predispose to aggressive tumors may help distinguish men who are more likely to discontinue AS. To investigate this, we undertook a multi-institutional genome-wide association study (GWAS) of 5,222 PC patients and 1,139 other patients from replication cohorts, all of whom initially elected AS and were followed over time for the potential outcome of conversion from AS to active treatment. In the GWAS we detected 18 variants associated with conversion, 15 of which were not previously associated with PC risk. With a transcriptome-wide association study (TWAS), we found two genes associated with conversion (MAST3, p = 6.9×10-7 and GAB2, p = 2.0×10-6). Moreover, increasing values of a previously validated 269-variant genetic risk score (GRS) for PC was positively associated with conversion (e.g., comparing the highest to the two middle deciles gave a hazard ratio [HR] = 1.13; 95% Confidence Interval [CI]= 0.94-1.36); whereas, decreasing values of a 36-variant GRS for prostate-specific antigen (PSA) levels were positively associated with conversion (e.g., comparing the lowest to the two middle deciles gave a HR = 1.25; 95% CI, 1.04-1.50). These results suggest that germline genetics may help inform and individualize the decision of AS-or the intensity of monitoring on AS-versus treatment for the initial management of patients with low-risk PC.
Collapse
Affiliation(s)
- Yu Jiang
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Travis J. Meyers
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Adaeze A. Emeka
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Lauren Folgosa Cooley
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Phillip R. Cooper
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Nicola Lancki
- Division of Biostatistics, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Irene Helenowski
- Division of Biostatistics, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Linda Kachuri
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Daniel W. Lin
- Fred Hutchinson Cancer Research Center, Cancer Prevention Program, Public Health Sciences, Seattle, WA 98109, USA
- Department of Urology, University of Washington, Seattle, WA 98195, USA
| | - Janet L. Stanford
- Fred Hutchinson Cancer Research Center, Cancer Epidemiology Program, Public Health Sciences, Seattle, WA 98109, USA
- Department of Epidemiology, University of Washington, School of Public Health, Seattle, WA 98195, USA
| | - Lisa F. Newcomb
- Fred Hutchinson Cancer Research Center, Cancer Prevention Program, Public Health Sciences, Seattle, WA 98109, USA
- Department of Urology, University of Washington, Seattle, WA 98195, USA
| | - Suzanne Kolb
- Fred Hutchinson Cancer Research Center, Cancer Epidemiology Program, Public Health Sciences, Seattle, WA 98109, USA
- Department of Epidemiology, University of Washington, School of Public Health, Seattle, WA 98195, USA
| | - Antonio Finelli
- Division of Urology, Department of Surgery, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Neil E. Fleshner
- Division of Urology, Department of Surgery, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Maria Komisarenko
- Division of Urology, Department of Surgery, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - James A. Eastham
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Behfar Ehdaie
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicole Benfante
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christopher J. Logothetis
- Departments of Genitourinary Medical Oncology and Urology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Justin R. Gregg
- Departments of Genitourinary Medical Oncology and Urology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cherie A. Perez
- Departments of Genitourinary Medical Oncology and Urology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sergio Garza
- Departments of Genitourinary Medical Oncology and Urology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeri Kim
- Departments of Genitourinary Medical Oncology and Urology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Leonard S. Marks
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Merdie Delfin
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Danielle Barsa
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Danny Vesprini
- Odette Cancer Centre, Sunnybrook Health and Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Laurence H. Klotz
- Odette Cancer Centre, Sunnybrook Health and Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Andrew Loblaw
- Odette Cancer Centre, Sunnybrook Health and Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Alexandre Mamedov
- Odette Cancer Centre, Sunnybrook Health and Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - S. Larry Goldenberg
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Celestia S. Higano
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Maria Spillane
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Eugenia Wu
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - H. Ballentine Carter
- Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christian P. Pavlovich
- Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mufaddal Mamawala
- Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tricia Landis
- Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter R. Carroll
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
| | - June M. Chan
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
| | - Matthew R. Cooperberg
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Janet E. Cowan
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
| | - Todd M. Morgan
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Javed Siddiqui
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Rabia Martin
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Eric A. Klein
- Glickman Urological and Kidney Institute, Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Karen Brittain
- Glickman Urological and Kidney Institute, Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Paige Gotwald
- Glickman Urological and Kidney Institute, Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Daniel A. Barocas
- Department of Urology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeremiah R. Dallmer
- Department of Urology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jennifer B. Gordetsky
- Department of Urology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Pam Steele
- Department of Urology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shilajit D. Kundu
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jazmine Stockdale
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Monique J. Roobol
- Department of Urology, Erasmus Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Lionne D.F. Venderbos
- Department of Urology, Erasmus Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Martin G. Sanda
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Rebecca Arnold
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Dattatraya Patil
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Christopher P. Evans
- Department of Urologic Surgery, University of California, Davis Medical Center, Sacramento, CA, USA
| | - Marc A. Dall’Era
- Department of Urologic Surgery, University of California, Davis Medical Center, Sacramento, CA, USA
| | - Anjali Vij
- Department of Urologic Surgery, University of California, Davis Medical Center, Sacramento, CA, USA
| | - Anthony J. Costello
- Department of Urology, Royal Melbourne Hospital and University of Melbourne, Melbourne, VIC, Australia
| | - Ken Chow
- Department of Urology, Royal Melbourne Hospital and University of Melbourne, Melbourne, VIC, Australia
| | - Niall M. Corcoran
- Department of Urology, Royal Melbourne Hospital and University of Melbourne, Melbourne, VIC, Australia
| | - Soroush Rais-Bahrami
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Courtney Phares
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Douglas S. Scherr
- Department of Urology, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY, USA
| | - Thomas Flynn
- Department of Urology, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY, USA
| | | | - Michael Koch
- Department of Urology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Courtney Rose Dhondt
- Department of Urology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joel B. Nelson
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dawn McBride
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michael S. Cookson
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kelly L. Stratton
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stephen Farriester
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Erin Hemken
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Tuula Pera
- University of Chicago Comprehensive Cancer Center, Chicago, IL, USA
| | | | | | | | - Stacy Loeb
- Departments of Urology and Population Health, New York University Langone Health and Manhattan Veterans Affairs Medical Center, New York, NY, USA
| | - Samir S. Taneja
- Departments of Urology and Population Health, New York University Langone Health and Manhattan Veterans Affairs Medical Center, New York, NY, USA
| | - Nataliya Byrne
- Departments of Urology and Population Health, New York University Langone Health and Manhattan Veterans Affairs Medical Center, New York, NY, USA
| | | | - Ann Martinez
- Department of Urology, Oregon Health and Science University, Portland, OR, USA
| | - Luc Boileau
- Department of Urology, Oregon Health and Science University, Portland, OR, USA
| | - Franklin D. Gaylis
- Genesis Healthcare Partners, Department of Urology, University of California, San Diego, CA, USA
| | | | - Nicholas Kirwen
- Division of Urology, NorthShore University Health System, Evanston, IL, USA
| | - Brian T. Helfand
- Division of Urology, NorthShore University Health System, Evanston, IL, USA
| | - Jianfeng Xu
- Division of Urology, NorthShore University Health System, Evanston, IL, USA
| | - Denise M. Scholtens
- Division of Biostatistics, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - William J. Catalona
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - John S. Witte
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Departments of Epidemiology and Population Health, Biomedical Data Science, and Genetics, Stanford University, Stanford, CA, USA
| |
Collapse
|
7
|
Wendeu-Foyet MG, Cénée S, Koudou Y, Trétarre B, Rébillard X, Cancel-Tassin G, Cussenot O, Boland A, Olaso R, Deleuze JF, Blanché H, Lamy PJ, Mulot C, Laurent-Puig P, Truong T, Menegaux F. Circadian genes polymorphisms, night work and prostate cancer risk: Findings from the EPICAP study. Int J Cancer 2020; 147:3119-3129. [PMID: 32506468 DOI: 10.1002/ijc.33139] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 05/04/2020] [Accepted: 05/18/2020] [Indexed: 11/10/2022]
Abstract
Over the past two decades, several studies have attempted to understand the hypothesis that disrupting the circadian rhythm may promote the development of cancer. Some have suggested that night work and some circadian genes polymorphisms are associated with cancer, including prostate cancer. Our study aims to test the hypothesis that prostate cancer risk among night workers may be modulated by genetic polymorphisms in the circadian pathway genes based on data from the EPICAP study, a population-based case-control study including 1511 men (732 cases/779 controls) with genotyped data. We estimated odds ratio (ORs) and P values of the association between prostate cancer and circadian gene variants using logistic regression models. We tested the interaction between circadian genes variants and night work indicators that were significantly associated with prostate cancer at pathway, gene and SNP levels. Analyses were also stratified by each of these night work indicators and by cancer aggressiveness. The circadian pathway was significantly associated with aggressive prostate cancer among night workers (P = .004), particularly for men who worked at night for <20 years (P = .0002) and those who performed long night shift (>10 hours, P = .001). At the gene level, we observed among night workers significant associations between aggressive prostate cancer and ARNTL, NPAS2 and RORA. At the SNP-level, no significant association was observed. Our findings provide some clues of a potential modulating effect of circadian genes in the relationship between night work and prostate cancer. Further investigation is warranted to confirm these findings and to better elucidate the biological pathways involved.
Collapse
Affiliation(s)
| | - Sylvie Cénée
- Université Paris-Saclay, UVSQ, Inserm, CESP, Villejuif, France
| | - Yves Koudou
- Université Paris-Saclay, UVSQ, Inserm, CESP, Villejuif, France
| | - Brigitte Trétarre
- Registre des Tumeurs de l'Hérault, EA 2415, ICM, Montpellier, France
| | | | - Géraldine Cancel-Tassin
- CeRePP, Hopital Tenon, Paris, France
- Sorbonne Université, GRC no. 5, ONCOTYPE-URO, AP-HP, Hôpital Tenon, Paris, France
| | - Olivier Cussenot
- CeRePP, Hopital Tenon, Paris, France
- Sorbonne Université, GRC no. 5, ONCOTYPE-URO, AP-HP, Hôpital Tenon, Paris, France
| | - Anne Boland
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Saint-Aubin, France
| | - Robert Olaso
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Saint-Aubin, France
| | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Saint-Aubin, France
| | - Hélène Blanché
- Centre d'Etude du Polymorphisme Humain (CEPH), Fondation Jean Dausset, Paris, France
| | - Pierre-Jean Lamy
- Clinique Beau Soleil, Service Urologie, Montpellier, France
- Institut médical d'Analyse Génomique-Imagenome, Labosud, Montpellier, France
| | - Claire Mulot
- Université Paris Descartes, INSERM UMR-S1147 EPIGENETEC, Paris, France
| | | | - Thérèse Truong
- Université Paris-Saclay, UVSQ, Inserm, CESP, Villejuif, France
| | | |
Collapse
|
8
|
He Z, Wang C, Xue H, Zhao R, Li G. Identification of a Metabolism-Related Risk Signature Associated With Clinical Prognosis in Glioblastoma Using Integrated Bioinformatic Analysis. Front Oncol 2020; 10:1631. [PMID: 33042807 PMCID: PMC7523182 DOI: 10.3389/fonc.2020.01631] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Altered metabolism of glucose, lipid and glutamine is a prominent hallmark of cancer cells. Currently, cell heterogeneity is believed to be the main cause of poor prognosis of glioblastoma (GBM) and is closely related to relapse caused by therapy resistance. However, the comprehensive model of genes related to glucose-, lipid- and glutamine-metabolism associated with the prognosis of GBM remains unclear, and the metabolic heterogeneity of GBM still needs to be further explored. Based on the expression profiles of 1,395 metabolism-related genes in three datasets of TCGA/CGGA/GSE, consistent cluster analysis revealed that GBM had three different metabolic status and prognostic clusters. Combining univariate Cox regression analysis and LASSO-penalized Cox regression machine learning methods, we identified a 17-metabolism-related genes risk signature associated with GBM prognosis. Kaplan-Meier analysis found that obtained signature could differentiate the prognosis of high- and low-risk patients in three datasets. Moreover, the multivariate Cox regression analysis and receiver operating characteristic curves indicated that the signature was an independent prognostic factor for GBM and had a strong predictive power. The above results were further validated in the CGGA and GSE13041 datasets, and consistent results were obtained. Gene set enrichment analysis (GSEA) suggested glycolysis gluconeogenesis and oxidative phosphorylation were significantly enriched in high- and low-risk GBM. Lastly Connectivity Map screened 54 potential compounds specific to different subgroups of GBM patients. Our study identified a novel metabolism-related gene signature, in addition the existence of three different metabolic status and two opposite biological processes in GBM were recognized, which revealed the metabolic heterogeneity of GBM. Robust metabolic subtypes and powerful risk prognostic models contributed a new perspective to the metabolic exploration of GBM.
Collapse
Affiliation(s)
- Zheng He
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Chengcheng Wang
- Department of Pharmacy, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Hao Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Rongrong Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| |
Collapse
|
9
|
Abstract
Circadian clocks are cell-autonomous self-sustaining oscillators that allow organisms to anticipate environmental changes throughout the solar day and persist in nearly every cell examined. Environmental or genetic disruption of circadian rhythms increases the risk of several types of cancer, but the underlying mechanisms are not well understood. Here, we discuss evidence connecting circadian rhythms-with emphasis on the cryptochrome proteins (CRY1/2)-to cancer through in vivo models, mechanisms involving known tumor suppressors and oncogenes, chemotherapeutic efficacy, and human cancer risk.
Collapse
Affiliation(s)
- Alanna B Chan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Katja A Lamia
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
10
|
Imada EL, Sanchez DF, Collado-Torres L, Wilks C, Matam T, Dinalankara W, Stupnikov A, Lobo-Pereira F, Yip CW, Yasuzawa K, Kondo N, Itoh M, Suzuki H, Kasukawa T, Hon CC, de Hoon MJL, Shin JW, Carninci P, Jaffe AE, Leek JT, Favorov A, Franco GR, Langmead B, Marchionni L. Recounting the FANTOM CAGE-Associated Transcriptome. Genome Res 2020; 30:1073-1081. [PMID: 32079618 PMCID: PMC7397872 DOI: 10.1101/gr.254656.119] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 02/11/2020] [Indexed: 02/02/2023]
Abstract
Long noncoding RNAs (lncRNAs) have emerged as key coordinators of biological and cellular processes. Characterizing lncRNA expression across cells and tissues is key to understanding their role in determining phenotypes, including human diseases. We present here FC-R2, a comprehensive expression atlas across a broadly defined human transcriptome, inclusive of over 109,000 coding and noncoding genes, as described in the FANTOM CAGE-Associated Transcriptome (FANTOM-CAT) study. This atlas greatly extends the gene annotation used in the original recount2 resource. We demonstrate the utility of the FC-R2 atlas by reproducing key findings from published large studies and by generating new results across normal and diseased human samples. In particular, we (a) identify tissue-specific transcription profiles for distinct classes of coding and noncoding genes, (b) perform differential expression analysis across thirteen cancer types, identifying novel noncoding genes potentially involved in tumor pathogenesis and progression, and (c) confirm the prognostic value for several enhancer lncRNAs expression in cancer. Our resource is instrumental for the systematic molecular characterization of lncRNA by the FANTOM6 Consortium. In conclusion, comprised of over 70,000 samples, the FC-R2 atlas will empower other researchers to investigate functions and biological roles of both known coding genes and novel lncRNAs.
Collapse
Affiliation(s)
- Eddie Luidy Imada
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21827, USA.,Departamento de Bioqúımica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Diego Fernando Sanchez
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21827, USA
| | | | - Christopher Wilks
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Tejasvi Matam
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21827, USA
| | - Wikum Dinalankara
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21827, USA
| | - Aleksey Stupnikov
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21827, USA
| | - Francisco Lobo-Pereira
- Departamento de Biologia General, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Chi-Wai Yip
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Kayoko Yasuzawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Naoto Kondo
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Masayoshi Itoh
- RIKEN, Preventive Medicine and Diagnostic Innovation Program, Yokohama, 351-0198, Japan
| | - Harukazu Suzuki
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Takeya Kasukawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Chung-Chau Hon
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | | | - Jay W Shin
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Piero Carninci
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Andrew E Jaffe
- Lieber Institute for Brain Development, Baltimore, Maryland 21205, USA.,Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205, USA.,Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
| | - Jeffrey T Leek
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
| | - Alexander Favorov
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21827, USA.,Laboratory of Systems Biology and Computational Genetics, VIGG RAS, 117971 Moscow, Russia
| | - Gloria R Franco
- Departamento de Bioqúımica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Ben Langmead
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA.,Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
| | - Luigi Marchionni
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21827, USA
| |
Collapse
|
11
|
Houlahan KE, Shiah YJ, Gusev A, Yuan J, Ahmed M, Shetty A, Ramanand SG, Yao CQ, Bell C, O'Connor E, Huang V, Fraser M, Heisler LE, Livingstone J, Yamaguchi TN, Rouette A, Foucal A, Espiritu SMG, Sinha A, Sam M, Timms L, Johns J, Wong A, Murison A, Orain M, Picard V, Hovington H, Bergeron A, Lacombe L, Lupien M, Fradet Y, Têtu B, McPherson JD, Pasaniuc B, Kislinger T, Chua MLK, Pomerantz MM, van der Kwast T, Freedman ML, Mani RS, He HH, Bristow RG, Boutros PC. Genome-wide germline correlates of the epigenetic landscape of prostate cancer. Nat Med 2019; 25:1615-1626. [PMID: 31591588 PMCID: PMC7418214 DOI: 10.1038/s41591-019-0579-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 08/13/2019] [Indexed: 12/16/2022]
Abstract
Oncogenesis is driven by germline, environmental and stochastic factors. It is unknown how these interact to produce the molecular phenotypes of tumors. We therefore quantified the influence of germline polymorphisms on the somatic epigenome of 589 localized prostate tumors. Predisposition risk loci influence a tumor's epigenome, uncovering a mechanism for cancer susceptibility. We identified and validated 1,178 loci associated with altered methylation in tumoral but not nonmalignant tissue. These tumor methylation quantitative trait loci influence chromatin structure, as well as RNA and protein abundance. One prominent tumor methylation quantitative trait locus is associated with AKT1 expression and is predictive of relapse after definitive local therapy in both discovery and validation cohorts. These data reveal intricate crosstalk between the germ line and the epigenome of primary tumors, which may help identify germline biomarkers of aggressive disease to aid patient triage and optimize the use of more invasive or expensive diagnostic assays.
Collapse
Affiliation(s)
- Kathleen E Houlahan
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Vector Institute, Toronto, Ontario, Canada
| | - Yu-Jia Shiah
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Alexander Gusev
- Division of Population Sciences, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jiapei Yuan
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Musaddeque Ahmed
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Anamay Shetty
- Division of Population Sciences, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- University of Cambridge, Cambridge, UK
| | - Susmita G Ramanand
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Cindy Q Yao
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Connor Bell
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Edward O'Connor
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Vincent Huang
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Michael Fraser
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | | | | | | | - Adrien Foucal
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | - Ankit Sinha
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Michelle Sam
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Lee Timms
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Jeremy Johns
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Ada Wong
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Alex Murison
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Michèle Orain
- Department of Pathology, Centre de recheche du CHU de Québec-Université Laval, Québec City, Québec, Canada
| | - Valérie Picard
- Division of Urology, Centre de recheche du CHU de Québec-Université Laval, Québec City, Québec, Canada
| | - Hélène Hovington
- Division of Urology, Centre de recheche du CHU de Québec-Université Laval, Québec City, Québec, Canada
| | - Alain Bergeron
- Division of Urology, Centre de recheche du CHU de Québec-Université Laval, Québec City, Québec, Canada
| | - Louis Lacombe
- Division of Urology, Centre de recheche du CHU de Québec-Université Laval, Québec City, Québec, Canada
| | - Mathieu Lupien
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Yves Fradet
- Division of Urology, Centre de recheche du CHU de Québec-Université Laval, Québec City, Québec, Canada
| | - Bernard Têtu
- Department of Pathology, Centre de recheche du CHU de Québec-Université Laval, Québec City, Québec, Canada
| | | | - Bogdan Pasaniuc
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Thomas Kislinger
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Melvin L K Chua
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Mark M Pomerantz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Theodorus van der Kwast
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada
| | - Matthew L Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- The Eli and Edythe L. Broad Institute, Cambridge, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ram S Mani
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Housheng H He
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Robert G Bristow
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada.
- Division of Cancer Sciences, Faculty of Biology, Health and Medicine, University of Manchester, Manchester, UK.
- The Christie NHS Foundation Trust, Manchester, UK.
- Cancer Research UK Manchester Institute, Manchester, UK.
- Manchester Cancer Research Centre, Manchester, UK.
| | - Paul C Boutros
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
- Vector Institute, Toronto, Ontario, Canada.
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
12
|
Morales-Santana S, Morell S, Leon J, Carazo-Gallego A, Jimenez-Lopez JC, Morell M. An Overview of the Polymorphisms of Circadian Genes Associated With Endocrine Cancer. Front Endocrinol (Lausanne) 2019; 10:104. [PMID: 30873119 PMCID: PMC6401647 DOI: 10.3389/fendo.2019.00104] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 02/04/2019] [Indexed: 12/17/2022] Open
Abstract
A major consequence of the world industrialized lifestyle is the increasing period of unnatural light in environments during the day and artificial lighting at night. This major change disrupts endogenous homeostasis with external circadian cues, which has been associated to higher risk of diseases affecting human health, mainly cancer among others. Circadian disruption promotes tumor development and accelerate its fast progression. The dysregulation mechanisms of circadian genes is greatly affected by the genetic variability of these genes. To date, several core circadian genes, also called circadian clock genes, have been identified, comprising the following: ARNTL, CLOCK, CRY1, CRY2, CSNK1E, NPAS2, NR1D1, NR1D2, PER1, PER2, PER3, RORA, and TIMELESS. The polymorphic variants of these circadian genes might contribute to an individual's risk to cancer. In this short review, we focused on clock circadian clock-related genes, major contributors of the susceptibility to endocrine-dependent cancers through affecting circadian clock, most likely affecting hormonal regulation. We examined polymorphisms affecting breast, prostate and ovarian carcinogenesis, in addition to pancreatic and thyroid cancer. Further study of the genetic composition in circadian clock-controlled tumors will be of great importance by establishing the foundation to discover novel genetic biomarkers for cancer prevention, prognosis and target therapies.
Collapse
Affiliation(s)
- Sonia Morales-Santana
- Proteomic Research Service, San Cecilio University Hospital, Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), Granada, Spain
- *Correspondence: Sonia Morales-Santana
| | - Santiago Morell
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
- Santiago Morell
| | - Josefa Leon
- Clinical Management Unit of Digestive Disease, San Cecilio University Hospital, Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), Granada, Spain
| | - Angel Carazo-Gallego
- Genomic Research Service, San Cecilio University Hospital, Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), Granada, Spain
| | - Jose C. Jimenez-Lopez
- Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, Spanish National Research Council (CSIC), Granada, Spain
- The UWA Institute of Agriculture and School of Agriculture and Environment, The University of Western Australia, Perth, WA, Australia
| | - María Morell
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada, Andalusian Regional Government, PTS Granada, Granada, Spain
| |
Collapse
|
13
|
Benna C, Rajendran S, Spiro G, Tropea S, Del Fiore P, Rossi CR, Mocellin S. Associations of clock genes polymorphisms with soft tissue sarcoma susceptibility and prognosis. J Transl Med 2018; 16:338. [PMID: 30518396 PMCID: PMC6280400 DOI: 10.1186/s12967-018-1715-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 11/30/2018] [Indexed: 12/28/2022] Open
Abstract
Background Dysfunction of the circadian clock and polymorphisms of some circadian genes have been linked to cancer development and progression. We investigated the relationship between circadian genes germline variation and susceptibility or prognosis of patients with soft tissue sarcoma. Patients and methods We considered the 14 single nucleotide polymorphisms (SNPs) of 6 core circadian genes that have a minor allele frequency > 5% and that are known to be associated with cancer risk or prognosis. Genotyping was performed by q-PCR. Peripheral blood and clinic-pathological data were available for 162 patients with liposarcoma or leiomyosarcoma and 610 healthy donors. Associations between the selected clock genes polymorphisms and sarcoma susceptibility or prognosis were tested assuming 3 models of inheritance: additive, recessive and dominant. Subgroup analysis based on sarcoma histotype was performed under the additive genetic model. Multivariate logistic regression and multivariate Cox proportional hazard regression analyses were utilized to assess the association between SNPs with patient susceptibility and survival, respectively. Pathway variation analysis was conducted employing the Adaptive Rank Truncated Product method. Results Six out of the 14 analyzed SNPs were statistically significantly associated with susceptibility or prognosis of soft tissue sarcoma (P < 0.05). The present analysis suggested that carriers of the minor allele of the CLOCK polymorphism rs1801260 (C) or of PER2 rs934945 (T) had a reduced predisposition to sarcoma (26% and 35% respectively with the additive model) and liposarcoma (33% and 41% respectively). The minor allele (A) of NPAS2 rs895520 was associated with an increased predisposition to sarcoma of 33% and leiomyosarcoma of 44%. RORA rs339972 C allele was associated with a decreased predisposition to develop sarcoma assuming an additive model (29%) and leiomyosarcoma (36%). PER1 rs3027178 was associated with a reduced predisposition only in liposarcoma subgroup (32%). rs7602358 located upstream PER2 was significantly associated with liposarcoma survival (HR: 1.98; 95% CI 1.02–3.85; P = 0.04). Germline genetic variation in the circadian pathway was associated with the risk of developing soft tissue sarcoma (P = 0.035). Conclusions Genetic variation of circadian genes appears to play a role in the determinism of patient susceptibility and prognosis. These findings prompt further studies to fully dissect the molecular mechanisms. Electronic supplementary material The online version of this article (10.1186/s12967-018-1715-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Clara Benna
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padua, Italy. .,Clinica Chirurgica I, Azienda Ospedaliera Padova, Padua, Italy.
| | | | - Giovanna Spiro
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Saveria Tropea
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padua, Italy.,Surgical Oncology Unit, Istituto Oncologico Veneto (IOV-IRCCS), Padua, Italy
| | - Paolo Del Fiore
- Surgical Oncology Unit, Istituto Oncologico Veneto (IOV-IRCCS), Padua, Italy
| | - Carlo Riccardo Rossi
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padua, Italy.,Surgical Oncology Unit, Istituto Oncologico Veneto (IOV-IRCCS), Padua, Italy
| | - Simone Mocellin
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padua, Italy.,Surgical Oncology Unit, Istituto Oncologico Veneto (IOV-IRCCS), Padua, Italy
| |
Collapse
|
14
|
Sheng W, Zhang H, Lu Y. Survival outcomes of locally advanced prostate cancer in patients aged < 50 years after local therapy in the contemporary US population. Int Urol Nephrol 2018; 50:1435-1444. [DOI: 10.1007/s11255-018-1931-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 07/03/2018] [Indexed: 12/19/2022]
|
15
|
Hu MB, Xu H, Hu JM, Zhu WH, Yang T, Jiang HW, Ding Q. Genetic polymorphisms in leptin, adiponectin and their receptors affect risk and aggressiveness of prostate cancer: evidence from a meta-analysis and pooled-review. Oncotarget 2018; 7:81049-81061. [PMID: 27768592 PMCID: PMC5348375 DOI: 10.18632/oncotarget.12747] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 10/12/2016] [Indexed: 01/04/2023] Open
Abstract
Leptin and adiponectin signaling was associated with development and progression of various cancers. The present study aimed to clarify the role of genetic variants in leptin, adiponectin and their receptors in prostate cancer. After comprehensive search and manuscript scanning, a total of 49 genetic variants were enrolled and examined for their relations to cancer risk and aggressiveness. In the meta-analysis, LEP rs7799039 (allele contrast: OR 1.133, 95%CI 1.024-1.254), ADIPOQ rs2241766 (allele contrast: OR 1.201, 95%CI 1.015-1.422) and ADIPOR1 rs10920531 (allele contrast: OR 1.184, 95%CI 1.075-1.305) variants were identified to be correlated with increased risk of prostate cancer. On the contrary, LEPR rs1137101 (allele contrast: OR 0.843, 95%CI 0.730-0.973) and ADIPOR1 rs2232853 (allele contrast: OR 0.638, 95%CI 0.535-0.760) variants were associated with decreased risk of prostate cancer. From the pooled-review, we additionally recognized eight variants associated with cancer risk and another eight variants associated with cancer aggressiveness, respectively. These observations indicated important roles of leptin, adiponectin and their receptors in the development and progression of prostate cancer. The identified polymorphisms might assist in developing better risk-assessment tools, as well as generating novel targeted therapies, especially for obese cancer patients with impaired leptin and adiponectin signaling.
Collapse
Affiliation(s)
- Meng-Bo Hu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Hua Xu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ji-Meng Hu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wen-Hui Zhu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Tian Yang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Hao-Wen Jiang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qiang Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
16
|
Dreussi E, Ecca F, Scarabel L, Gagno S, Toffoli G. Immunogenetics of prostate cancer: a still unexplored field of study. Pharmacogenomics 2018; 19:263-283. [PMID: 29325503 DOI: 10.2217/pgs-2017-0163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The immune system is a double-edged sword with regard to the prostate cancer (PCa) battle. Immunogenetics, the study of the potential role of immune-related polymorphisms, is taking its first steps in the treatment of this malignancy. This review summarizes the most recent papers addressing the potential of immunogenetics in PCa, reporting immune-related polymorphisms associated with tumor aggressiveness, treatment toxicity and patients' prognosis. With some peculiarities, RNASEL, IL-6, IL-10, IL-1β and MMP7 have arisen as the most significant biomarkers in PCa treatment and management, having a potential clinical role. Validation prospective clinical studies are required to translate immunogenetics into precision treatment of PCa.
Collapse
Affiliation(s)
- Eva Dreussi
- Department of Experimental & Clinical Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, 33081, Italy
| | - Fabrizio Ecca
- Department of Experimental & Clinical Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, 33081, Italy
| | - Lucia Scarabel
- Department of Experimental & Clinical Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, 33081, Italy
| | - Sara Gagno
- Department of Experimental & Clinical Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, 33081, Italy
| | - Giuseppe Toffoli
- Department of Experimental & Clinical Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, 33081, Italy
| |
Collapse
|
17
|
Germline variants in IL4, MGMT and AKT1 are associated with prostate cancer-specific mortality: An analysis of 12,082 prostate cancer cases. Prostate Cancer Prostatic Dis 2018; 21:228-237. [PMID: 29298992 PMCID: PMC6026113 DOI: 10.1038/s41391-017-0029-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 11/09/2017] [Accepted: 11/20/2017] [Indexed: 02/08/2023]
Abstract
Background Prostate cancer (PCa) is a leading cause of mortality and genetic factors can influence tumour aggressiveness. Several germline variants have been associated with PCa-specific mortality (PCSM), but further replication evidence is needed. Methods Twenty-two previously identified PCSM-associated genetic variants were genotyped in seven PCa cohorts (12,082 patients; 1544 PCa deaths). For each cohort, Cox proportional hazards models were used to calculate hazard ratios and 95% confidence intervals for risk of PCSM associated with each variant. Data were then combined using a meta-analysis approach. Results Fifteen SNPs were associated with PCSM in at least one of the seven cohorts. In the meta-analysis, after adjustment for clinicopathological factors, variants in the MGMT (rs2308327; HR 0.90; p-value = 3.5 × 10−2) and IL4 (rs2070874; HR 1.22; p-value = 1.1 × 10−3) genes were confirmed to be associated with risk of PCSM. In analyses limited to men diagnosed with local or regional stage disease, a variant in AKT1, rs2494750, was also confirmed to be associated with PCSM risk (HR 0.81; p-value = 3.6 × 10−2). Conclusions This meta-analysis confirms the association of three genetic variants with risk of PCSM, providing further evidence that genetic background plays a role in PCa-specific survival. While these variants alone are not sufficient as prognostic biomarkers, these results may provide insights into the biological pathways modulating tumour aggressiveness.
Collapse
|
18
|
Laviana AA, Reisz PA, Resnick MJ. Prostate Cancer Screening in African-American Men. Prostate Cancer 2018. [DOI: 10.1007/978-3-319-78646-9_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
19
|
Szulkin R, Clements MS, Magnusson PKE, Wiklund FE, Kuja-Halkola R. Estimating Heritability of Prostate Cancer-Specific Survival Using Population-Based Registers. Prostate 2017; 77:900-907. [PMID: 28247425 DOI: 10.1002/pros.23344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 02/13/2017] [Indexed: 11/07/2022]
Abstract
BACKGROUND There is a strong genetic component in prostate cancer development with an estimated heritability of 58%. In addition, recent epidemiological assessments show a familial component in prostate cancer-specific survival, which could be due to either common genetics or environment. In this study we sought to estimate the heritability of prostate cancer-specific survival by studying brothers and father-son pairs in Sweden. METHODS We used linkage records from three Swedish national registers: the Multi-Generation Register, the Cancer Register, and the Cause of Death Register. One thousand seven hundred twenty-eight brother pairs and 6,444 father-son pairs, where both family members were diagnosed with prostate cancer, were followed for prostate cancer mortality. By assuming that (i) brothers on average share 50% of their segregating alleles and 100% environment and (ii) fathers and sons share 50% of their segregating alleles and no environment, we implemented a model including influences of additive genetics (heritability), shared environment and non-shared environment for survival data. A conditional likelihood estimation procedure was developed to fit the model. Data simulation was applied to validate model assumptions. RESULTS In a model that adjusted for age at diagnosis and calendar period, the estimated heritability of prostate cancer-specific survival was 0.10 (95% CI = 0.00-0.20) that was borderline significantly different from zero (P = 0.057). The shared environment component was not significantly different from zero with a point estimate of 0.00 (95% CI = 0.00-0.13). Simulation studies and sensitivity analysis revealed that the estimated heritability component was robust, whereas the shared environmental component may be underestimated. CONCLUSIONS Heritability of prostate cancer-specific survival is considerably lower than for prostate cancer incidence. This supports a hypothesis that susceptibility of disease and progression of disease are separate mechanisms that involve different genes. Further assessment of the genetic basis of prostate cancer-specific survival is warranted. Prostate 77:900-907, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Robert Szulkin
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Division of Family Medicine, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Huddinge, Sweden
| | - Mark S Clements
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik E Wiklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ralf Kuja-Halkola
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
20
|
Li H, Lu YF, Chen H, Liu J. Dysregulation of metallothionein and circadian genes in human hepatocellular carcinoma. Chronobiol Int 2016; 34:192-202. [PMID: 27997226 DOI: 10.1080/07420528.2016.1256300] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 10/26/2016] [Accepted: 10/31/2016] [Indexed: 12/25/2022]
Abstract
Hepatocellular carcinoma (HCC) is the major threat to human health, and disruption of circadian clock genes is implicated in hepatocarcinogenesis. This study examined the dysregulation of metallothioneins and circadian genes in achieved human HCC (n = 24), peri-HCC tissues (n = 24) as compared with normal human livers (n = 36). Total RNA was extracted and reverse transcribed. Real-time RT-qPCR was performed to determine the expression of genes of interest. The results demonstrated the downregulation of metallothionein-1 (MT-1), MT-2, and metal transcription factor-1 (MFT-1) in human HCC as compared with Peri-HCC and normal tissues. MTs are a biomarker for HCC and have typical circadian rhythms; the expression of major circadian clock genes was also determined. HCC produced a dramatic decrease in the expression of core clock genes, circadian locomotor output cycles kaput (Clock) and brain and muscle Arnt-like protein 1 (Bmal1), and decreased the expression of the clock feedback control genes, Periods (Per1, Per2) and Cryptochromes (Cry1, Cry2). On the other hand, the expression of clock target genes nuclear orphan receptor factor protein (Nr1d1) and D-box-binding protein (Dbp) was upregulated as compared with Peri-HCC and normal livers. Peri-HCC also had mild alterations in these gene expressions. In summary, the present study clearly demonstrated the dysregulation of MTs and circadian clock genes in human HCC, which could provide the information of targeting MT and circadian clock in HCC management.
Collapse
Affiliation(s)
- Huan Li
- a Key Lab for Basic Pharmacology of Ministry of Education , Zunyi Medical College , Zunyi , China
| | - Yuan-Fu Lu
- a Key Lab for Basic Pharmacology of Ministry of Education , Zunyi Medical College , Zunyi , China
| | - Hong Chen
- b The Institute of Organ Transplantation , The General Hospital of Chinese People's Armed Police Forces , Beijing , China
| | - Jie Liu
- a Key Lab for Basic Pharmacology of Ministry of Education , Zunyi Medical College , Zunyi , China
| |
Collapse
|
21
|
Alshaker H, Sacco K, Alfraidi A, Muhammad A, Winkler M, Pchejetski D. Leptin signalling, obesity and prostate cancer: molecular and clinical perspective on the old dilemma. Oncotarget 2016; 6:35556-63. [PMID: 26376613 PMCID: PMC4742124 DOI: 10.18632/oncotarget.5574] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/16/2015] [Indexed: 01/22/2023] Open
Abstract
The prevalence of global obesity is increasing. Obesity is associated with general cancer-related morbidity and mortality and is a known risk factor for development of specific cancers. A recent large systematic review of 24 studies based on meta-analysis of 11,149 patients with prostate cancer showed a significant correlation between obesity and the risk of advanced prostate cancer. Further, a sustained reduction in BMI correlates with a decreased risk of developing aggressive disease. On the other hand, the correlation between consuming different products and prostate cancer occurrence/risk is limited.Here, we review the role of adipose tissue from an endocrine perspective and outline the effect of adipokines on cancer metabolism, with particular focus on leptin. Leptin exerts its physiological and pathological effects through modification of intracellular signalling, most notably activating the Janus kinase (JAK) 2/signal transducer and activator of transcription (STAT) 3 pathway and recently shown sphingolipid pathway. Both high levels of leptin in circulation and leptin receptor mutation are associated with prostate cancer risk in human patients; however, the in vivo mechanistic evidence is less conclusive.Given the complexity of metabolic cancer pathways, it is possible that leptin may have varying effects on prostate cancer at different stages of its development, a point that may be addressed by further epidemiological studies.
Collapse
Affiliation(s)
- Heba Alshaker
- Department of Surgery and Cancer, Imperial College London, London, UK.,Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, Petra University, Amman, Jordan
| | - Keith Sacco
- University of Malta Medical School, Mater Dei Hospital, Tal-Qroqq, MSD, Malta
| | - Albandri Alfraidi
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Aun Muhammad
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Mathias Winkler
- Department of Surgery and Cancer, Imperial College London, London, UK
| | | |
Collapse
|
22
|
Börnigen D, Tyekucheva S, Wang X, Rider JR, Lee GS, Mucci LA, Sweeney C, Huttenhower C. Computational Reconstruction of NFκB Pathway Interaction Mechanisms during Prostate Cancer. PLoS Comput Biol 2016; 12:e1004820. [PMID: 27078000 PMCID: PMC4831844 DOI: 10.1371/journal.pcbi.1004820] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 02/19/2016] [Indexed: 12/21/2022] Open
Abstract
Molecular research in cancer is one of the largest areas of bioinformatic investigation, but it remains a challenge to understand biomolecular mechanisms in cancer-related pathways from high-throughput genomic data. This includes the Nuclear-factor-kappa-B (NFκB) pathway, which is central to the inflammatory response and cell proliferation in prostate cancer development and progression. Despite close scrutiny and a deep understanding of many of its members’ biomolecular activities, the current list of pathway members and a systems-level understanding of their interactions remains incomplete. Here, we provide the first steps toward computational reconstruction of interaction mechanisms of the NFκB pathway in prostate cancer. We identified novel roles for ATF3, CXCL2, DUSP5, JUNB, NEDD9, SELE, TRIB1, and ZFP36 in this pathway, in addition to new mechanistic interactions between these genes and 10 known NFκB pathway members. A newly predicted interaction between NEDD9 and ZFP36 in particular was validated by co-immunoprecipitation, as was NEDD9's potential biological role in prostate cancer cell growth regulation. We combined 651 gene expression datasets with 1.4M gene product interactions to predict the inclusion of 40 additional genes in the pathway. Molecular mechanisms of interaction among pathway members were inferred using recent advances in Bayesian data integration to simultaneously provide information specific to biological contexts and individual biomolecular activities, resulting in a total of 112 interactions in the fully reconstructed NFκB pathway: 13 (11%) previously known, 29 (26%) supported by existing literature, and 70 (63%) novel. This method is generalizable to other tissue types, cancers, and organisms, and this new information about the NFκB pathway will allow us to further understand prostate cancer and to develop more effective prevention and treatment strategies. In molecular research in cancer it remains challenging to uncover biomolecular mechanisms in cancer-related pathways from high-throughput genomic data, including the Nuclear-factor-kappa-B (NFκB) pathway. Despite close scrutiny and a deep understanding of many of the NFκB pathway members’ biomolecular activities, the current list of pathway members and a systems-level understanding of their interactions remains incomplete. In this study, we provide the first steps toward computational reconstruction of interaction mechanisms of the NFκB pathway in prostate cancer. We identified novel roles for 8 genes in this pathway and new mechanistic interactions between these genes and 10 known pathway members. We combined 651 gene expression datasets with 1.4M interactions to predict the inclusion of 40 additional genes in the pathway. Molecular mechanisms of interaction were inferred using recent advances in Bayesian data integration to simultaneously provide information specific to biological contexts and individual biomolecular activities, resulting in 112 interactions in the fully reconstructed NFκB pathway. This method is generalizable, and this new information about the NFκB pathway will allow us to further understand prostate cancer.
Collapse
Affiliation(s)
- Daniela Börnigen
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, United States of America.,The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Svitlana Tyekucheva
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Xiaodong Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jennifer R Rider
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, United States of America
| | - Gwo-Shu Lee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, United States of America
| | - Christopher Sweeney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, United States of America.,The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| |
Collapse
|
23
|
Hurley PJ, Sundi D, Shinder B, Simons BW, Hughes RM, Miller RM, Benzon B, Faraj SF, Netto GJ, Vergara IA, Erho N, Davicioni E, Karnes RJ, Yan G, Ewing C, Isaacs SD, Berman DM, Rider JR, Jordahl KM, Mucci LA, Huang J, An SS, Park BH, Isaacs WB, Marchionni L, Ross AE, Schaeffer EM. Germline Variants in Asporin Vary by Race, Modulate the Tumor Microenvironment, and Are Differentially Associated with Metastatic Prostate Cancer. Clin Cancer Res 2015; 22:448-58. [PMID: 26446945 DOI: 10.1158/1078-0432.ccr-15-0256] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 09/10/2015] [Indexed: 12/20/2022]
Abstract
PURPOSE Prostate cancers incite tremendous morbidity upon metastatic growth. We previously identified Asporin (ASPN) as a potential mediator of metastatic progression found within the tumor microenvironment. ASPN contains an aspartic acid (D)-repeat domain and germline polymorphisms in D-repeat-length have been associated with degenerative diseases. Associations of germline ASPN D polymorphisms with risk of prostate cancer progression to metastatic disease have not been assessed. EXPERIMENTAL DESIGN Germline ASPN D-repeat-length was retrospectively analyzed in 1,600 men who underwent radical prostatectomy for clinically localized prostate cancer and in 548 noncancer controls. Multivariable Cox proportional hazards models were used to test the associations of ASPN variations with risk of subsequent oncologic outcomes, including metastasis. Orthotopic xenografts were used to establish allele- and stroma-specific roles for ASPN D variants in metastatic prostate cancer. RESULTS Variation at the ASPN D locus was differentially associated with poorer oncologic outcomes. ASPN D14 [HR, 1.72; 95% confidence interval (CI), 1.05-2.81, P = 0.032] and heterozygosity for ASPN D13/14 (HR, 1.86; 95% CI, 1.03-3.35, P = 0.040) were significantly associated with metastatic recurrence, while homozygosity for the ASPN D13 variant was significantly associated with a reduced risk of metastatic recurrence (HR, 0.44; 95% CI, 0.21-0.94, P = 0.035) in multivariable analyses. Orthotopic xenografts established biologic roles for ASPN D14 and ASPN D13 variants in metastatic prostate cancer progression that were consistent with patient-based data. CONCLUSIONS We observed associations between ASPN D variants and oncologic outcomes, including metastasis. Our data suggest that ASPN expressed in the tumor microenvironment is a heritable modulator of metastatic progression.
Collapse
Affiliation(s)
- Paula J Hurley
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland. Department of Oncology, Johns Hopkins University, Baltimore, Maryland. Sidney Kimmel Comprehensive Cancer Institute, Johns Hopkins University, Baltimore, Maryland.
| | - Debasish Sundi
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - Brian Shinder
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - Brian W Simons
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland. Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, Maryland
| | - Robert M Hughes
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - Rebecca M Miller
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - Benjamin Benzon
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - Sheila F Faraj
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - George J Netto
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | | | - Nicholas Erho
- Genome Dx Biosciences Inc., Vancouver, British Columbia, Canada
| | - Elai Davicioni
- Genome Dx Biosciences Inc., Vancouver, British Columbia, Canada
| | | | - Guifang Yan
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - Charles Ewing
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - Sarah D Isaacs
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - David M Berman
- Department of Pathology and Molecular Medicine and Cancer Research Institute, Queens University, Kingston, Ontario, Canada
| | - Jennifer R Rider
- Department of Epidemiology, Harvard University, T.H. Chan School of Public Health, Boston, Massachusetts
| | - Kristina M Jordahl
- Department of Epidemiology, Harvard University, T.H. Chan School of Public Health, Boston, Massachusetts
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard University, T.H. Chan School of Public Health, Boston, Massachusetts
| | - Jessie Huang
- The Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Steven S An
- The Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland. The Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland. Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, Maryland
| | - Ben H Park
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland. Sidney Kimmel Comprehensive Cancer Institute, Johns Hopkins University, Baltimore, Maryland
| | - William B Isaacs
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - Luigi Marchionni
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Ashley E Ross
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland. Department of Oncology, Johns Hopkins University, Baltimore, Maryland. Sidney Kimmel Comprehensive Cancer Institute, Johns Hopkins University, Baltimore, Maryland. Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Edward M Schaeffer
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland. Department of Oncology, Johns Hopkins University, Baltimore, Maryland. Sidney Kimmel Comprehensive Cancer Institute, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
24
|
Replication of a genetic variant for prostate cancer-specific mortality. Prostate Cancer Prostatic Dis 2015; 18:260-3. [PMID: 25939514 PMCID: PMC4537383 DOI: 10.1038/pcan.2015.18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/02/2015] [Accepted: 02/28/2015] [Indexed: 11/09/2022]
Abstract
BACKGROUND Few genetic variants have been confirmed as being associated with prostate cancer-specific mortality (PCSM). A recent study identified 22 candidate single-nucleotide polymorphisms (SNPs) associated with PCSM in a Seattle-based patient cohort. Five of these associations were replicated in an independent Swedish cohort. METHODS We genotyped these 22 SNPs in Physicians' Health Study (PHS) participants diagnosed with prostate cancer (PCa). Using the same model that was found to be most significant in the Seattle cohort, we examined the association of these SNPs with lethal disease with Cox proportional hazards models. RESULTS One SNP, rs5993891 in the ARVCF gene on chromosome 22q11, which had also replicated in the Swedish cohort, was also significantly associated with PCSM in the PHS cohort (hazard ratio (HR)=0.32; P=0.01). When we tested this SNP in an additional cohort (Health Professionals Follow-up Study, HPFS), the association was null (HR=0.95, P=0.90); however, a meta-analysis across all studies showed a statistically significant association with a HR of 0.52 (0.29-0.93, P=0.03). CONCLUSIONS The association of rs5993891 with PCSM was further replicated in PHS and remains significant in a meta-analysis, though there was no association in HPFS. This SNP may contribute to a genetic panel of SNPs to determine at diagnosis whether a patient is more likely to exhibit an indolent or aggressive form of PCa. This study also emphasizes the importance of multiple rounds of replication.
Collapse
|
25
|
Cheng J, Ondracek RP, Mehedint DC, Kasza KA, Xu B, Gill S, Azabdaftari G, Yao S, Morrison CD, Mohler JL, Marshall JR. Association of fatty-acid synthase polymorphisms and expression with outcomes after radical prostatectomy. Prostate Cancer Prostatic Dis 2015; 18:182-9. [PMID: 25868764 DOI: 10.1038/pcan.2015.11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 02/24/2015] [Accepted: 02/28/2015] [Indexed: 11/09/2022]
Abstract
BACKGROUND Fatty-acid synthase (FASN), selectively overexpressed in prostate cancer (PCa) cells, has been described as linked to the aggressiveness of PCa. Constitutional genetic variation of the FASN gene and the expression levels of FASN protein in cancer cells could thus be expected to predict outcome after radical prostatectomy (RP). This study evaluates the associations of malignant tissue status, neoadjuvant androgen deprivation therapy (NADT) and single-nucleotide polymorphisms (SNPs) of FASN with FASN protein expression in prostate tissue. The study then examines the associations of FASN SNPs and gene expression with three measures of post-prostatectomy outcome. METHODS Seven tagging FASN SNPs were genotyped in 659 European American men who underwent RP at Roswell Park Cancer Institute between 1993 and 2005. FASN protein expression was assessed using immunohistochemistry. The patients were followed for an average of 6.9 years (range: 0.1-20.6 years). Outcome was assessed using three end points: biochemical failure, treatment failure and development of distant metastatic PCa. Cox proportional hazards analyses were used to evaluate the associations of the tagging SNPs and FASN expression with these end points. Bivariate associations with outcomes were considered; the associations also were controlled for known aggressiveness indicators. RESULTS Overall, no SNPs were associated with any known aggressiveness indicators. FASN staining intensity was stronger in malignant than in benign tissue, and NADT was associated with decreased FASN staining in both benign and malignant tissue. The relationships of FASN SNPs and staining intensity with outcome were less clear. One SNP, rs4246444, showed a weak association with outcome. FASN staining intensity also showed a weak and seemingly contradictory relationship with outcome. CONCLUSIONS Additional study with longer follow-up and populations that include more metastatic patients is warranted.
Collapse
Affiliation(s)
- J Cheng
- 1] Department of Cancer Prevention and Population Science, Roswell Park Cancer Institute, Buffalo, NY, USA [2] Department of Pathology, University at Buffalo, Buffalo, NY, USA
| | - R P Ondracek
- Department of Cancer Prevention and Population Science, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - D C Mehedint
- Department of Urology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - K A Kasza
- Department of Cancer Prevention and Population Science, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - B Xu
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - S Gill
- Department of Pathology, University at Buffalo, Buffalo, NY, USA
| | - G Azabdaftari
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - S Yao
- Department of Cancer Prevention and Population Science, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - C D Morrison
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - J L Mohler
- Department of Urology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - J R Marshall
- Department of Cancer Prevention and Population Science, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
26
|
Helfand BT, Roehl KA, Cooper PR, McGuire BB, Fitzgerald LM, Cancel-Tassin G, Cornu JN, Bauer S, Van Blarigan EL, Chen X, Duggan D, Ostrander EA, Gwo-Shu M, Zhang ZF, Chang SC, Jeong S, Fontham ETH, Smith G, Mohler JL, Berndt SI, McDonnell SK, Kittles R, Rybicki BA, Freedman M, Kantoff PW, Pomerantz M, Breyer JP, Smith JR, Rebbeck TR, Mercola D, Isaacs WB, Wiklund F, Cussenot O, Thibodeau SN, Schaid DJ, Cannon-Albright L, Cooney KA, Chanock SJ, Stanford JL, Chan JM, Witte J, Xu J, Bensen JT, Taylor JA, Catalona WJ. Associations of prostate cancer risk variants with disease aggressiveness: results of the NCI-SPORE Genetics Working Group analysis of 18,343 cases. Hum Genet 2015; 134:439-50. [PMID: 25715684 PMCID: PMC4586077 DOI: 10.1007/s00439-015-1534-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 02/06/2015] [Indexed: 01/18/2023]
Abstract
Genetic studies have identified single nucleotide polymorphisms (SNPs) associated with the risk of prostate cancer (PC). It remains unclear whether such genetic variants are associated with disease aggressiveness. The NCI-SPORE Genetics Working Group retrospectively collected clinicopathologic information and genotype data for 36 SNPs which at the time had been validated to be associated with PC risk from 25,674 cases with PC. Cases were grouped according to race, Gleason score (Gleason ≤ 6, 7, ≥ 8) and aggressiveness (non-aggressive, intermediate, and aggressive disease). Statistical analyses were used to compare the frequency of the SNPs between different disease cohorts. After adjusting for multiple testing, only PC-risk SNP rs2735839 (G) was significantly and inversely associated with aggressive (OR = 0.77; 95 % CI 0.69-0.87) and high-grade disease (OR = 0.77; 95 % CI 0.68-0.86) in European men. Similar associations with aggressive (OR = 0.72; 95 % CI 0.58-0.89) and high-grade disease (OR = 0.69; 95 % CI 0.54-0.87) were documented in African-American subjects. The G allele of rs2735839 was associated with disease aggressiveness even at low PSA levels (<4.0 ng/mL) in both European and African-American men. Our results provide further support that a PC-risk SNP rs2735839 near the KLK3 gene on chromosome 19q13 may be associated with aggressive and high-grade PC. Future prospectively designed, case-case GWAS are needed to identify additional SNPs associated with PC aggressiveness.
Collapse
Affiliation(s)
- Brian T Helfand
- Department of Surgery, Division of Urology, John and Carol Walter Center for Urological Health, NorthShore University Health System, Evanston, IL, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Zhang D, Tang N, Liu Y, Wang EH. ARVCF expression is significantly correlated with the malignant phenotype of non-small cell lung cancer. Mol Carcinog 2015; 54 Suppl 1:E185-91. [PMID: 25683624 DOI: 10.1002/mc.22281] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 12/08/2014] [Accepted: 12/12/2014] [Indexed: 01/14/2023]
Abstract
Armadillo repeat gene deleted in velo-cardio-facial syndrome (ARVCF) is a member of the p120 catenin (p120ctn) family; it contains nine central Armadillo repeats and binds to the juxtamembrane domain of E-cadherin. We used immunohistochemistry to measure ARVCF expression in 121 patients with NSCLC and western blotting to examine differences in ARVCF expression between lung cancer and adjacent normal lung tissues. We interfered with ARVCF expression in two lung cancer cell lines and measured its effects on invasion and proliferation. ARVCF expression correlated with the malignant phenotype and poor prognosis. We also observed ARVCF-dependent changes in small GTPase (mainly RhoA) activity in lung cancer cells. We confirmed that ARVCF plays an important role in the malignant phenotype.
Collapse
Affiliation(s)
- Di Zhang
- The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, China
| | - Na Tang
- The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, China
| | - Yang Liu
- The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, China
| | - En-Hua Wang
- The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, China
| |
Collapse
|
28
|
Luo Y, Ye Z, Li K, Chen R, Li S, Pang J. Associations between polymorphisms in the IL-4 and IL-4 receptor genes and urinary carcinomas: a meta-analysis. Int J Clin Exp Med 2015; 8:1227-1233. [PMID: 25785117 PMCID: PMC4358572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 01/13/2015] [Indexed: 06/04/2023]
Abstract
UNLABELLED To evaluate the association between polymorphisms of interleukin-4 (IL-4) and IL-4 receptor (IL-4R) genes and risk of renal cell cancer (RCC), bladder cancer (BC), and prostate cancer (PC) based on meta-analysis. PubMed, Web of Science and SpecilalSCI(TM) were searched for studies published up to May 2014 that reported the association between IL-4 or IL-4R and RCC, BC or PC risk. Odds ratio (OR)/Hazard ratio (HR) and 95% confidence interval (CI) were analyzed to evaluate the association. Meta-analysis showed that the IL-4R polymorphism rs1805010 was associated with increased RCC risk (CC/CT vs. TT: OR=1.266, 95% CI 1.09-1.472, P=0.002). The IL-4 haplotypes, IL4-589T and IL4-33T, were associated with higher survival rate of the patients comparted with the haplotype IL-4-589C-33C (P<0.05). The IL-4 polymorphism rs2243250 was associated with an increased risk of developing multiple BCs (OR=2.52, P=0.033). The IL-4 polymorphisms rs2243228, rs2243250, and rs22272480 were significantly associated with PC risk (rs2243228 CC vs. CA/AA OR=0.27, 95% CI 0.09-0.84, P=0.03; rs2243350 TT vs. CT/CC OR=2.16, 95% CI 1.06-4.40, P=0.03, CC vs. CT/TT OR=1.31, 95% CI 1.05-1.65, P=0.02; rs2227284 TT vs. GT/GG OR=1.98, 95% CI 1.30-3.00, P=0.001). In addition, IL-4 polymorphism rs2070874 was associated with PC mortality. Three polymorphisms (rs2070874, rs1805015, and rs1801275) were not associated with RCC, BC, and PC. The IL-4R polymorphism rs1805015 might be associated with RCC risk. IL-4 rs2243250 carriers had increased risk of developing multiple BCs. IL-4 polymorphisms rs2243228, rs2243250, rs2227284, and rs2070874 were associated with PC risk or mortality.
Collapse
Affiliation(s)
- Yun Luo
- Department of Urology, The Third Affiliated Hospital, Sun Yat-Sen UniversityGuangzhou 510630, China
| | - Zhiqiang Ye
- Department of Emergency Medicine, The Third Affiliated Hospital, Sun Yat-Sen UniversityGuangzhou 510630, China
| | - Ke Li
- Department of Urology, The Third Affiliated Hospital, Sun Yat-Sen UniversityGuangzhou 510630, China
| | - Ruihan Chen
- Department of Emergency Medicine, The Third Affiliated Hospital, Sun Yat-Sen UniversityGuangzhou 510630, China
| | - Shigeng Li
- Department of Emergency Medicine, The Third Affiliated Hospital, Sun Yat-Sen UniversityGuangzhou 510630, China
| | - Jun Pang
- Department of Urology, The Third Affiliated Hospital, Sun Yat-Sen UniversityGuangzhou 510630, China
| |
Collapse
|
29
|
Marcello MA, Calixto AR, de Almeida JFM, Martins MB, Cunha LL, Cavalari CAA, Etchebehere ECS, da Assumpção LVM, Geloneze B, Carvalho AL, Ward LS. Polymorphism in LEP and LEPR May Modify Leptin Levels and Represent Risk Factors for Thyroid Cancer. Int J Endocrinol 2015; 2015:173218. [PMID: 25810718 PMCID: PMC4355553 DOI: 10.1155/2015/173218] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 02/13/2015] [Indexed: 12/19/2022] Open
Abstract
Purpose. To understand the role of polymorphisms in the LEP (rs7799039 and rs2167270) and LEPR (rs1137101 and rs1137100) genes in DTC susceptibility and their effect on leptin levels. Methods. We studied 153 patients with DTC and 234 controls through TaqMan SNP Genotyping and ELISA, comparing these data to the clinicopathological data of patients with DTC. Results. Patients with AA genotype of rs7799039 had higher levels of serum leptin (9.22 ± 0.98 ng/mL) than those with AG genotype (10.07 ± 0.60 ng/mL; P = 0.005). Individuals with AG genotype of rs2167270 also produced higher serum leptin levels (10.05 ± 0.59 ng/mL) than the subjects with GG genotype (9.52 ± 0.79 ng/mL; P < 0.05). A multivariate logistic regression adjusted for gender, age, and BMI showed that the AG genotype of rs7799039 was an independent risk for DTC (OR, 11.689; P = 0.0183; 95% CI, 1.516-90.119). Similarly, AG and GG genotypes of rs1137101 increased the susceptibility to DTC (OR, 3.747; P = 0.027; 95% CI, 1.161-12.092 and OR, 5.437; P = 0.013; 95% CI, 1.426-20.729). Conclusions. We demonstrated that rs7799039 and rs2167270 polymorphisms modify the serum leptin concentrations in patients with DTC. Furthermore, polymorphisms rs7799039 and rs1137101 increase the risk of DTC development, although they do not correlate with tumor aggressiveness.
Collapse
Affiliation(s)
- Marjory Alana Marcello
- Laboratory of Cancer Molecular Genetics (Gemoca), Faculty of Medical Sciences, University of Campinas (FCM-Unicamp), Rua Tessalia Vieira de Camargo 126, 13083-970 Campinas, SP, Brazil
- *Marjory Alana Marcello:
| | - Antonio Ramos Calixto
- Laboratory of Investigation on Metabolism and Diabetes (LIMED), Faculty of Medical Sciences, University of Campinas (FCM-Unicamp), Rua Carlos Chagas 420, 13083-878 Campinas, SP, Brazil
| | - Jacqueline Fatima Martins de Almeida
- Laboratory of Cancer Molecular Genetics (Gemoca), Faculty of Medical Sciences, University of Campinas (FCM-Unicamp), Rua Tessalia Vieira de Camargo 126, 13083-970 Campinas, SP, Brazil
| | - Mariana Bonjiorno Martins
- Laboratory of Cancer Molecular Genetics (Gemoca), Faculty of Medical Sciences, University of Campinas (FCM-Unicamp), Rua Tessalia Vieira de Camargo 126, 13083-970 Campinas, SP, Brazil
| | - Lucas Leite Cunha
- Laboratory of Cancer Molecular Genetics (Gemoca), Faculty of Medical Sciences, University of Campinas (FCM-Unicamp), Rua Tessalia Vieira de Camargo 126, 13083-970 Campinas, SP, Brazil
| | - Camila Ayume Amano Cavalari
- Laboratory of Cancer Molecular Genetics (Gemoca), Faculty of Medical Sciences, University of Campinas (FCM-Unicamp), Rua Tessalia Vieira de Camargo 126, 13083-970 Campinas, SP, Brazil
| | - Elba C. S. Etchebehere
- Service of Nuclear Medicine, University of Campinas, Rua Vital Brasil 251, 13083-888 Campinas, SP, Brazil
| | | | - Bruno Geloneze
- Laboratory of Investigation on Metabolism and Diabetes (LIMED), Faculty of Medical Sciences, University of Campinas (FCM-Unicamp), Rua Carlos Chagas 420, 13083-878 Campinas, SP, Brazil
| | - Andre Lopes Carvalho
- Department of Head and Neck Surgery, Barretos Cancer Hospital, Rua Antenor Duarte Vilela 1331, 14784-400 Barretos, SP, Brazil
| | - Laura Sterian Ward
- Laboratory of Cancer Molecular Genetics (Gemoca), Faculty of Medical Sciences, University of Campinas (FCM-Unicamp), Rua Tessalia Vieira de Camargo 126, 13083-970 Campinas, SP, Brazil
| |
Collapse
|
30
|
Markt SC, Valdimarsdottir UA, Shui IM, Sigurdardottir LG, Rider JR, Tamimi RM, Batista JL, Haneuse S, Flynn-Evans E, Lockley SW, Czeisler CA, Stampfer MJ, Launer L, Harris T, Smith AV, Gudnason V, Lindstrom S, Kraft P, Mucci LA. Circadian clock genes and risk of fatal prostate cancer. Cancer Causes Control 2015; 26:25-33. [PMID: 25388799 PMCID: PMC4282953 DOI: 10.1007/s10552-014-0478-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 10/09/2014] [Indexed: 01/20/2023]
Abstract
PURPOSE Circadian genes may be involved in regulating cancer-related pathways, including cell proliferation, DNA damage response, and apoptosis. We aimed to assess the role of genetic variation in core circadian rhythm genes with the risk of fatal prostate cancer and first morning void urinary 6-sulfatoxymelatonin levels. METHODS We used unconditional logistic regression to evaluate the association of 96 single-nucleotide polymorphisms (SNPs) across 12 circadian-related genes with fatal prostate cancer in the AGES-Reykjavik cohort (n = 24 cases), the Health Professionals Follow-Up Study (HPFS) (n = 40 cases), and the Physicians' Health Study (PHS) (n = 105 cases). We used linear regression to evaluate the association between SNPs and first morning void urinary 6-sulfatoxymelatonin levels in AGES-Reykjavik. We used a kernel machine test to evaluate whether multimarker SNP sets in the pathway (gene based) were associated with our outcomes. RESULTS None of the individual SNPs were consistently associated with fatal prostate cancer across the three cohorts. In each cohort, gene-based analyses showed that variation in the CRY1 gene was nominally associated with fatal prostate cancer (p values = 0.01, 0.01, and 0.05 for AGES-Reykjavik, HPFS, and PHS, respectively). In AGES-Reykjavik, SNPs in TIMELESS (four SNPs), NPAS2 (six SNPs), PER3 (two SNPs) and CSNK1E (one SNP) were nominally associated with 6-sulfatoxymelatonin levels. CONCLUSION We did not find a strong and consistent association between variation in core circadian clock genes and fatal prostate cancer risk, but observed nominally significant gene-based associations with fatal prostate cancer and 6-sulfatoxymelatonin levels.
Collapse
Affiliation(s)
- Sarah C Markt
- Department of Epidemiology, Harvard School of Public Health, 677 Huntington Avenue, Boston, MA, 02115-6018, USA,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Karyadi DM, Zhao S, He Q, McIntosh L, Wright JL, Ostrander EA, Feng Z, Stanford JL. Confirmation of genetic variants associated with lethal prostate cancer in a cohort of men from hereditary prostate cancer families. Int J Cancer 2014; 136:2166-71. [PMID: 25273821 DOI: 10.1002/ijc.29241] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 08/11/2014] [Accepted: 08/26/2014] [Indexed: 12/27/2022]
Abstract
Germline genetic variants have been suggested as prognostic biomarkers for identifying patients at high risk for lethal prostate cancer (PCa). Validation studies have confirmed the association of several single nucleotide polymorphisms (SNPs) with fatal PCa, but whether these variants affect PCa-specific mortality (PCSM) in patients with an inherited predisposition to PCa, based on familial history, is unknown. For this study, a cohort of 957 PCa patients from 270 hereditary prostate cancer families of European ancestry was genotyped for a panel of 22 PCSM-associated SNPs. Death certificates were reviewed to confirm cause of death. Mixed-effect Cox proportional hazards models were used to assess survival according to genotypes, accounting for relatedness and clinicopathological factors. Within this cohort, 98 PCa deaths were confirmed over an average follow-up period of 12.7 years after diagnosis. Variant allele carriers for three SNPs had significantly altered risk for PCSM [rs635261 at RNASEL, hazard ratio (HR), 0.35, 95% CI, 0.18-0.66; p = 0.002; rs915927 in XRCC1, HR, 1.91, 95% CI, 1.21-3.02; p = 0.009; and rs2494750 at AKT1, HR, 0.45, 95% CI, 0.23-0.90; p = 0.016). These results confirm the association of genetic variation in three genes with PCa lethality in a cohort of men with an inherited susceptibility to the disease and provide validation evidence that germline SNPs provide prognostic information for PCa patients. Development of a panel of germline biomarkers with clinical utility for distinguishing patients at detection who have an increased risk for fatal PCa is warranted.
Collapse
Affiliation(s)
- Danielle M Karyadi
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Nikolić ZZ, Pavićević DLS, Romac SP, Brajušković GN. Genetic variants within endothelial nitric oxide synthase gene and prostate cancer: a meta-analysis. Clin Transl Sci 2014; 8:23-31. [PMID: 25164276 DOI: 10.1111/cts.12203] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Several variants within gene-encoding endothelial isoform of nitric oxide synthase have been reported to confer prostate cancer (PCa) susceptibility and/or progression. Nevertheless, studies referring to this issue have yielded inconsistent results. In order to elucidate the involvement of these variants in prostate carcinogenesis, we have conducted a meta-analysis of previously published case-control and relevant case-only studies. Eleven studies comprising in total 3,806 cases and 4,466 controls were included in the meta-analysis which yielded evidence of association of rs2070744 (ORCC = 1.43, 95% CI 1.04-1.97; p = 0.03) and intron 4a/b variant (ORab+aa = 1.47, 95% CI 1.00-2.14; p = 0.05) with PCa risk under recessive and dominant model, respectively. Furthermore, PCa patients carrying 4a/b a allele were found to have an increased risk of cancer progression to a less differentiated form, characterized by a high Gleason score (OR = 2.29, 95% CI 1.51-3.49; p < 0.01) and to higher TNM stage (OR = 2.55, 95% CI 1.71-3.81; p < 0.01). These results support the involvement of NOS3 variants in molecular pathogenesis of PCa.
Collapse
|
33
|
Demichelis F, Stanford JL. Genetic predisposition to prostate cancer: Update and future perspectives. Urol Oncol 2014; 33:75-84. [PMID: 24996773 DOI: 10.1016/j.urolonc.2014.04.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 04/27/2014] [Accepted: 04/28/2014] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Prostate cancer is the second most frequent cancer in men worldwide and kills over 250,000 men worldwide every year. Prostate cancer is a heterogeneous disease at the clinical and the molecular level. The Scandinavian Twin Registry Study demonstrated that in contrast to most malignancies where environment was the overriding influence, heritable factors account for more than fifty percent of prostate cancers. METHODS AND MATERIALS We review the literature on prostate cancer risk variants (rare and common) including SNPs and Copy Number Variants (CNVs) and discuss the potential implications of significant variants for prostate cancer patient care. RESULTS The search for prostate cancer susceptibility genes has included both family-based studies and case-control studies utilizing a variety of approaches from array-based to sequencing-based studies. A major challenge is to identify genetic variants associated with more aggressive, potentially lethal prostate cancer and to understand their role in the progression of the disease. CONCLUSION Future risk models useful in the clinical setting will likely incorporate several risk loci rather than single variants and may be dependent on an individual patient's ethnic background.
Collapse
Affiliation(s)
- Francesca Demichelis
- Centre for Integrative Biology, University of Trento, Trento, Italy; Institute for Computational Biomedicine, Weill Medical College of Cornell University, New York, NY; Institute for Precision Medicine, Weill Medical College of Cornell University and New York Presbyterian Hospital, New York, NY.
| | - Janet L Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA
| |
Collapse
|
34
|
Xue X, Liu F, Han Y, Li P, Yuan B, Wang X, Chen Y, Kuang Y, Zhi Q, Zhao H. Silencing NPAS2 promotes cell growth and invasion in DLD-1 cells and correlated with poor prognosis of colorectal cancer. Biochem Biophys Res Commun 2014; 450:1058-62. [PMID: 24978311 DOI: 10.1016/j.bbrc.2014.06.104] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 06/22/2014] [Indexed: 02/07/2023]
Abstract
Emerging evidences show that circadian rhythm disorder is an important factor of tumor initiation and development. Neuronal PAS domain protein2 (NPAS2), which is the largest circadian gene, has been proved to be a novel prognostic biomarker in breast cancer and non-Hodgkin's lymphoma. However, the potential functions of NPAS2 in colorectal cancer are still unknown. In our present study, we detected the mRNA expressions of NPAS2 in 108 CRC patients by RT-PCR, and found that NPAS2 expression was significantly down-regulated in tumor tissues than that in NATs. Clinicopathologic analysis revealed that low expression of NPAS2 was associated with the tumor size, TNM stage and tumor distance metastasis in colorectal cancer (p<0.05). Furthermore, we effectively down-regulated NPAS2 mRNA expression by transfecting RNA interfere fragments into DLD-1 cells, and our results in vitro demonstrated that silencing NPAS2 expression could promote cell proliferation, cell invasion and increase the wound healing ability (p<0.05). However, down-regulating NPAS2 expression did not influence the apoptotic rate in DLD-1 cells (p>0.05). In conclusion, our study suggested that NPAS2, functioned as a potential tumor suppressor gene, could serve as a promising target and potential prognostic indicator for colorectal cancer.
Collapse
Affiliation(s)
- Xiaofeng Xue
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Fei Liu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Ye Han
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Pu Li
- Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Department of Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Bin Yuan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xu Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yan Chen
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yuting Kuang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Qiaoming Zhi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Hong Zhao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| |
Collapse
|
35
|
Cao Y, Lindström S, Schumacher F, Stevens VL, Albanes D, Berndt S, Boeing H, Bueno-de-Mesquita HB, Canzian F, Chamosa S, Chanock SJ, Diver WR, Gapstur SM, Gaziano JM, Giovannucci EL, Haiman CA, Henderson B, Johansson M, Le Marchand L, Palli D, Rosner B, Siddiq A, Stampfer M, Stram DO, Tamimi R, Travis RC, Trichopoulos D, Willett WC, Yeager M, Kraft P, Hsing AW, Pollak M, Lin X, Ma J. Insulin-like growth factor pathway genetic polymorphisms, circulating IGF1 and IGFBP3, and prostate cancer survival. J Natl Cancer Inst 2014; 106:dju085. [PMID: 24824313 PMCID: PMC4081624 DOI: 10.1093/jnci/dju085] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 03/03/2014] [Accepted: 03/04/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The insulin-like growth factor (IGF) signaling pathway has been implicated in prostate cancer (PCa) initiation, but its role in progression remains unknown. METHODS Among 5887 PCa patients (704 PCa deaths) of European ancestry from seven cohorts in the National Cancer Institute Breast and Prostate Cancer Cohort Consortium, we conducted Cox kernel machine pathway analysis to evaluate whether 530 tagging single nucleotide polymorphisms (SNPs) in 26 IGF pathway-related genes were collectively associated with PCa mortality. We also conducted SNP-specific analysis using stratified Cox models adjusting for multiple testing. In 2424 patients (313 PCa deaths), we evaluated the association of prediagnostic circulating IGF1 and IGFBP3 levels and PCa mortality. All statistical tests were two-sided. RESULTS The IGF signaling pathway was associated with PCa mortality (P = .03), and IGF2-AS and SSTR2 were the main contributors (both P = .04). In SNP-specific analysis, 36 SNPs were associated with PCa mortality with P trend less than .05, but only three SNPs in the IGF2-AS remained statistically significant after gene-based corrections. Two were in linkage disequilibrium (r 2 = 1 for rs1004446 and rs3741211), whereas the third, rs4366464, was independent (r 2 = 0.03). The hazard ratios (HRs) per each additional risk allele were 1.19 (95% confidence interval [CI] = 1.06 to 1.34; P trend = .003) for rs3741211 and 1.44 (95% CI = 1.20 to 1.73; P trend < .001) for rs4366464. rs4366464 remained statistically significant after correction for all SNPs (P trend.corr = .04). Prediagnostic IGF1 (HRhighest vs lowest quartile = 0.71; 95% CI = 0.48 to 1.04) and IGFBP3 (HR = 0.93; 95% CI = 0.65 to 1.34) levels were not associated with PCa mortality. CONCLUSIONS The IGF signaling pathway, primarily IGF2-AS and SSTR2 genes, may be important in PCa survival.
Collapse
|
36
|
Abstract
Humans as diurnal beings are active during the day and rest at night. This daily oscillation of behavior and physiology is driven by an endogenous circadian clock not environmental cues. In modern societies, changes in lifestyle have led to a frequent disruption of the endogenous circadian homeostasis leading to increased risk of various diseases including cancer. The clock is operated by the feedback loops of circadian genes and controls daily physiology by coupling cell proliferation and metabolism, DNA damage repair, and apoptosis in peripheral tissues with physical activity, energy homeostasis, immune and neuroendocrine functions at the organismal level. Recent studies have revealed that defects in circadian genes due to targeted gene ablation in animal models or single nucleotide polymorphism, deletion, deregulation and/or epigenetic silencing in humans are closely associated with increased risk of cancer. In addition, disruption of circadian rhythm can disrupt the molecular clock in peripheral tissues in the absence of circadian gene mutations. Circadian disruption has recently been recognized as an independent cancer risk factor. Further study of the mechanism of clock-controlled tumor suppression will have a significant impact on human health by improving the efficiencies of cancer prevention and treatment.
Collapse
Affiliation(s)
- Nicole M Kettner
- Department of Pediatrics/U.S. Department of Agriculture/Agricultural Research Service/ Children's Nutrition Research Center, Baylor College of Medicine , Houston, TX , USA
| | | | | |
Collapse
|
37
|
Salinas CA, Tsodikov A, Ishak-Howard M, Cooney KA. Prostate cancer in young men: an important clinical entity. Nat Rev Urol 2014; 11:317-23. [PMID: 24818853 PMCID: PMC4191828 DOI: 10.1038/nrurol.2014.91] [Citation(s) in RCA: 194] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Prostate cancer is considered a disease of older men (aged >65 years), but today over 10% of new diagnoses in the USA occur in young men aged ≤55 years. Early-onset prostate cancer, that is prostate cancer diagnosed at age ≤55 years, differs from prostate cancer diagnosed at an older age in several ways. Firstly, among men with high-grade and advanced-stage prostate cancer, those diagnosed at a young age have a higher cause-specific mortality than men diagnosed at an older age, except those over age 80 years. This finding suggests that important biological differences exist between early-onset prostate cancer and late-onset disease. Secondly, early-onset prostate cancer has a strong genetic component, which indicates that young men with prostate cancer could benefit from evaluation of genetic risk. Furthermore, although the majority of men with early-onset prostate cancer are diagnosed with low-risk disease, the extended life expectancy of these patients exposes them to long-term effects of treatment-related morbidities and to long-term risk of disease progression leading to death from prostate cancer. For these reasons, patients with early-onset prostate cancer pose unique challenges, as well as opportunities, for both research and clinical communities. Current data suggest that early-onset prostate cancer is a distinct phenotype-from both an aetiological and clinical perspective-that deserves further attention.
Collapse
Affiliation(s)
- Claudia A. Salinas
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Alex Tsodikov
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Miriam Ishak-Howard
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Kathleen A. Cooney
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Urology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
38
|
Insulin-like Growth Factor Pathway Genetic Polymorphisms, Circulating IGF1 and IGFBP3, and Prostate Cancer Survival. J Natl Cancer Inst 2014; 106:dju218. [PMCID: PMC4111284 DOI: 10.1093/jnci/dju218] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 03/03/2014] [Accepted: 03/04/2014] [Indexed: 04/11/2024] Open
Abstract
Background The insulin-like growth factor (IGF) signaling pathway has been implicated in prostate cancer (PCa) initiation, but its role in progression remains unknown. Methods Among 5887 PCa patients (704 PCa deaths) of European ancestry from seven cohorts in the National Cancer Institute Breast and Prostate Cancer Cohort Consortium, we conducted Cox kernel machine pathway analysis to evaluate whether 530 tagging single nucleotide polymorphisms (SNPs) in 26 IGF pathway-related genes were collectively associated with PCa mortality. We also conducted SNP-specific analysis using stratified Cox models adjusting for multiple testing. In 2424 patients (313 PCa deaths), we evaluated the association of prediagnostic circulating IGF1 and IGFBP3 levels and PCa mortality. All statistical tests were two-sided. Results The IGF signaling pathway was associated with PCa mortality (P = .03), and IGF2-AS and SSTR2 were the main contributors (both P = .04). In SNP-specific analysis, 36 SNPs were associated with PCa mortality with P trend less than .05, but only three SNPs in the IGF2-AS remained statistically significant after gene-based corrections. Two were in linkage disequilibrium (r 2 = 1 for rs1004446 and rs3741211), whereas the third, rs4366464, was independent (r 2 = 0.03). The hazard ratios (HRs) per each additional risk allele were 1.19 (95% confidence interval [CI] = 1.06 to 1.34; P trend = .003) for rs3741211 and 1.44 (95% CI = 1.20 to 1.73; P trend < .001) for rs4366464. rs4366464 remained statistically significant after correction for all SNPs (P trend.corr = .04). Prediagnostic IGF1 (HRhighest vs lowest quartile = 0.71; 95% CI = 0.48 to 1.04) and IGFBP3 (HR = 0.93; 95% CI = 0.65 to 1.34) levels were not associated with PCa mortality. Conclusions The IGF signaling pathway, primarily IGF2-AS and SSTR2 genes, may be important in PCa survival.
Collapse
|
39
|
Helfand BT, Catalona WJ. The Epidemiology and Clinical Implications of Genetic Variation in Prostate Cancer. Urol Clin North Am 2014; 41:277-97. [DOI: 10.1016/j.ucl.2014.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
40
|
Lévesque É, Laverdière I, Audet-Walsh É, Caron P, Rouleau M, Fradet Y, Lacombe L, Guillemette C. Steroidogenic Germline Polymorphism Predictors of Prostate Cancer Progression in the Estradiol Pathway. Clin Cancer Res 2014; 20:2971-83. [DOI: 10.1158/1078-0432.ccr-13-2567] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
41
|
Madden MH, Anic GM, Thompson RC, Nabors LB, Olson JJ, Browning JE, Monteiro AN, Egan KM. Circadian pathway genes in relation to glioma risk and outcome. Cancer Causes Control 2014; 25:25-32. [PMID: 24135790 PMCID: PMC3947318 DOI: 10.1007/s10552-013-0305-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 10/08/2013] [Indexed: 12/11/2022]
Abstract
PURPOSE There is growing evidence that circadian disruption may alter risk and aggressiveness of cancer. We evaluated common genetic variants in the circadian gene pathway for associations with glioma risk and patient outcome in a US clinic-based case-control study. METHODS Subjects were genotyped for 17 candidate single nucleotide polymorphisms in ARNTL, CRY1, CRY2, CSNK1E, KLHL30, NPAS2, PER1, PER3, CLOCK, and MYRIP. Unconditional logistic regression was used to estimate age and gender-adjusted odds ratios (OR) and 95 % confidence intervals (CI) for glioma risk under three inheritance models (additive, dominant, and recessive). Proportional hazards regression was used to estimate hazard ratios for glioma-related death among 441 patients with high-grade tumors. Survival associations were validated using The Cancer Genome Atlas (TCGA) dataset. RESULTS A variant in PER1 (rs2289591) was significantly associated with overall glioma risk (per variant allele OR 0.80; 95 % CI 0.66-0.97; p trend = 0.027). The variant allele for CLOCK rs11133391 under a recessive model increased risk of oligodendroglioma (OR 2.41; 95 % CI 1.31-4.42; p = 0.005), though not other glioma subtypes (p for heterogeneity = 0.0033). The association remained significant after false discovery rate adjustment (p = 0.008). Differential associations by gender were observed for MYRIP rs6599077 and CSNK1E rs1534891 though differences were not significant after adjustment for multiple testing. No consistent mortality associations were identified. Several of the examined genes exhibited differential expression in glioblastoma multiforme versus normal brain in TCGA data (MYRIP, ARNTL, CRY1, KLHL30, PER1, CLOCK, and PER3), and expression of NPAS2 was significantly associated with a poor patient outcome in TCGA patients. CONCLUSION This exploratory analysis provides some evidence supporting a role for circadian genes in the onset of glioma and possibly the outcome of glioma.
Collapse
Affiliation(s)
- Melissa H. Madden
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa FL 33612, USA
| | - Gabriella M. Anic
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa FL 33612, USA
| | - Reid C. Thompson
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - L. Burton Nabors
- Neuro-oncology Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeffrey J. Olson
- Department of Neurosurgery, Emory School of Medicine, Atlanta, GA 30322, USA
| | - James E. Browning
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa FL 33612, USA
| | - Alvaro N. Monteiro
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa FL 33612, USA
| | - Kathleen M. Egan
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa FL 33612, USA
| |
Collapse
|
42
|
Jones DZ, Ragin C, Kidd NC, Flores-Obando RE, Jackson M, McFarlane-Anderson N, Tulloch-Reid M, Kimbro KS, Kidd LR. The impact of genetic variants in inflammatory-related genes on prostate cancer risk among men of African Descent: a case control study. Hered Cancer Clin Pract 2013; 11:19. [PMID: 24359571 PMCID: PMC3929257 DOI: 10.1186/1897-4287-11-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 12/03/2013] [Indexed: 01/13/2023] Open
Abstract
Purpose Although case–control studies have evaluated the role of variant inflammatory-related loci in prostate cancer, their impact is virtually unknown among men of African descent. To address this, we evaluated the impact of inflammatory cytokine single nucleotide polymorphisms (SNPs) on prostate cancer risk for men of African descent. Methods Forty-four SNPs in inflammatory cytokine-associated genes were evaluated among 814 African-American and Jamaican men (279 prostate cancer cases and 535 controls) using Illumina’s Golden gate genotyping system. Individual SNP effects were evaluated using logistic regression analysis. Results Four SNPs were modestly associated with prostate cancer after adjusting for age. In the total population, inheritance of the IL1R2 rs11886877 AA, IL8RB rs11574752 AA, TNF rs1800629 GA + AA, and TNF rs673 GA genotypes modestly increased prostate cancer risk by 1.45 to 11.7-fold relative to the referent genotype. Among U.S. men, age-adjusted dominant, recessive and additive genetic models for the IL1R2 rs11886877 locus were linked to an increase in prostate cancer susceptibility. However, these main effects did not persist after adjusting for multiple hypothesis testing. Conclusion Our preliminary data does not strongly support the hypothesis that inflammatory-related sequence variants influence prostate cancer risk among men of African descent. However, further evaluation is needed to assess whether other variant inflammatory-related genes may contribute to prostate cancer risk and disease progression in larger and ethnically diverse multi-center studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Lacreis R Kidd
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
43
|
Tsuchiya N, Matsui S, Narita S, Kamba T, Mitsuzuka K, Hatakeyama S, Horikawa Y, Inoue T, Saito S, Ohyama C, Arai Y, Ogawa O, Habuchi T. Distinct cancer-specific survival in metastatic prostate cancer patients classified by a panel of single nucleotide polymorphisms of cancer-associated genes. Genes Cancer 2013; 4:54-60. [PMID: 23946871 DOI: 10.1177/1947601913481354] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Accepted: 02/10/2013] [Indexed: 12/20/2022] Open
Abstract
Individual genetic variations may have a significant influence on the survival of metastatic prostate cancer (PCa) patients. We aimed to identify target genes and their variations involved in the survival of PCa patients using a single nucleotide polymorphism (SNP) panel. A total of 185 PCa patients with bone metastasis at the initial diagnosis were analyzed. Germline DNA in each patient was genotyped using a cancer SNP panel that contained 1,421 SNPs in 408 cancer-related genes. SNPs associated with survival were screened by a log-rank test. Fourteen SNPs in 6 genes, XRCC4, PMS1, GATA3, IL13, CASP8, and IGF1, were identified to have a statistically significant association with cancer-specific survival. The cancer-specific survival times of patients grouped according to the number of risk genotypes of 6 SNPs selected from the 14 SNPs differed significantly (0-1 v. 2-3 v. 4-6 risk genotypes; P = 7.20 × 10(-8)). The high-risk group was independently associated with survival in a multivariate analysis that included conventional clinicopathological variables (P = 0.0060). We identified 14 candidate SNPs in 6 cancer-related genes, which were associated with poor survival in patients with metastatic PCa. A panel of SNPs may help predict the survival of those patients.
Collapse
Affiliation(s)
- Norihiko Tsuchiya
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ohlander A, Zilio C, Hammerle T, Zelenin S, Klink G, Chiari M, Bock K, Russom A. Genotyping of single nucleotide polymorphisms by melting curve analysis using thin film semi-transparent heaters integrated in a lab-on-foil system. LAB ON A CHIP 2013; 13:2075-2082. [PMID: 23592049 DOI: 10.1039/c3lc50171j] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The recent technological advances in micro/nanotechnology present new opportunities to combine microfluidics with microarray technology for the development of small, sensitive, single-use, point-of-care molecular diagnostic devices. As such, the integration of microarray and plastic microfluidic systems is an attractive low-cost alternative to glass based microarray systems. This paper presents the integration of a DNA microarray and an all-polymer microfluidic foil system with integrated thin film heaters, which demonstrate DNA analysis based on melting curve analysis (MCA). A novel micro-heater concept using semi-transparent copper heaters manufactured by roll-to-roll and lift-off on polyethylene naphthalate (PEN) foil has been developed. Using a mesh structure, heater surfaces have been realized in only one single metallization step, providing more efficient and homogenous heating characteristics than conventional meander heaters. A robust DNA microarray spotting protocol was adapted on Parylene C coated heater-foils, using co-polymer poly(DMA-NAS-MAPS) to enable covalent immobilization of DNA. The heaters were integrated in a microfluidic channel using lamination foils and MCA of the spotted DNA duplexes showed single based discrimination of mismatched over matched target DNA-probes. Finally, as a proof of principle, we perform MCA on PCR products to detect the Leu7Pro polymorphism of the neutropeptide Y related to increased risk of Type II diabetes, BMI and depression.
Collapse
Affiliation(s)
- Anna Ohlander
- Fraunhofer EMFT, Hansastrasse 27d, 80686 Munich, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Keyes M, Macaulay C, Hayes M, Korbelik J, Morris WJ, Palcic B. DNA ploidy measured on archived pretreatment biopsy material may correlate with prostate-specific antigen recurrence after prostate brachytherapy. Int J Radiat Oncol Biol Phys 2013; 86:829-34. [PMID: 23688814 DOI: 10.1016/j.ijrobp.2013.04.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 04/03/2013] [Accepted: 04/08/2013] [Indexed: 10/26/2022]
Abstract
PURPOSE To explore whether DNA ploidy of prostate cancer cells determined from archived transrectal ultrasound-guided biopsy specimens correlates with disease-free survival. METHODS AND MATERIALS Forty-seven failures and 47 controls were selected from 1006 consecutive low- and intermediate-risk patients treated with prostate (125)I brachytherapy (July 1998-October 2003). Median follow-up was 7.5 years. Ten-year actuarial disease-free survival was 94.1%. Controls were matched using age, initial prostate-specific antigen level, clinical stage, Gleason score, use of hormone therapy, and follow-up (all P nonsignificant). Seventy-eight specimens were successfully processed; 27 control and 20 failure specimens contained more than 100 tumor cells were used for the final analysis. The Feulgen-Thionin stained cytology samples from archived paraffin blocks were used to determine the DNA ploidy of each tumor by measuring integrated optical densities. RESULTS The samples were divided into diploid and aneuploid tumors. Aneuploid tumors were found in 16 of 20 of the failures (80%) and 8 of 27 controls (30%). Diploid DNA patients had a significantly lower rate of disease recurrence (P=.0086) (hazard ratio [HR] 0.256). On multivariable analysis, patients with aneuploid tumors had a higher prostate-specific antigen failure rate (HR 5.13). Additionally, those with "excellent" dosimetry (V100 >90%; D90 >144 Gy) had a significantly lower recurrence rate (HR 0.25). All patients with aneuploid tumors and dosimetry classified as "nonexcellent" (V100 <90%; D90 <144 Gy) (5 of 5) had disease recurrence, compared with 40% of patients with aneuploid tumors and "excellent" dosimetry (8 of 15). In contrast, dosimetry did not affect the outcome for diploid patients. CONCLUSIONS Using core biopsy material from archived paraffin blocks, DNA ploidy correctly classified the majority of failures and nonfailures in this study. The results suggest that DNA ploidy can be used as a useful marker for aggressiveness of localized prostate cancer. A larger study will be necessary to further confirm our hypothesis.
Collapse
Affiliation(s)
- Mira Keyes
- Radiation Oncology, Provincial Prostate Brachytherapy Program, Vancouver Cancer Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
| | | | | | | | | | | |
Collapse
|
46
|
Tsuchiya N, Narita S, Inoue T, Saito M, Numakura K, Huang M, Hatakeyama S, Satoh S, Saito S, Ohyama C, Arai Y, Ogawa O, Habuchi T. Insulin-like growth factor-1 genotypes and haplotypes influence the survival of prostate cancer patients with bone metastasis at initial diagnosis. BMC Cancer 2013; 13:150. [PMID: 23530598 PMCID: PMC3622563 DOI: 10.1186/1471-2407-13-150] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 03/18/2013] [Indexed: 12/01/2022] Open
Abstract
Background The insulin-like growth factor-1 (IGF-1) plays an important role in growth of prostate cancer (PCa) cells and facilitating the development and progression of PCa. This study aimed to evaluate the association of polymorphisms in three linkage disequilibrium (LD) blocks of the IGF-1 on the survival of metastatic PCa patients. Methods A total of 215 patients with bone metastases at initial presentation were included in this study. The cytosine-adenine (CA) repeat polymorphism and rs12423791 were selected as representative polymorphisms in the LD blocks 1 and 2, respectively. Haplotype in the LD block 3 was analyzed using two tag single nucleotide polymorphisms (SNPs), rs6220 and rs7136446. Cancer-specific survival rate was estimated from the Kaplan-Meier curve, and the survival data were compared using the log-rank test. Results Cancer-specific survival was significantly associated with the CA repeat polymorphism, rs12423791, and rs6220 (P = 0.013, 0.014, and 0.014, respectively). Although rs7136446 had no significant association with survival, the haplotype in the LD block 3 was significantly associated with cancer-specific survival (P = 0.0003). When the sum of the risk genetic factors in each LD block (19-repeat allele, C allele of rs12423791, or C-T haplotype) was considered, patients with all the risk factors had significantly shorter cancer specific-survival than those with 0–2 risk factors (P = 0.0003). Conclusions Polymorphisms in the IGF-1, especially a haplotype in the LD block 3, are assumed to be genetic markers predicting the outcome of metastatic PCa.
Collapse
Affiliation(s)
- Norihiko Tsuchiya
- Department of Urology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Amin Al Olama A, Kote-Jarai Z, Schumacher FR, Wiklund F, Berndt SI, Benlloch S, Giles GG, Severi G, Neal DE, Hamdy FC, Donovan JL, Hunter DJ, Henderson BE, Thun MJ, Gaziano M, Giovannucci EL, Siddiq A, Travis RC, Cox DG, Canzian F, Riboli E, Key TJ, Andriole G, Albanes D, Hayes RB, Schleutker J, Auvinen A, Tammela TL, Weischer M, Stanford JL, Ostrander EA, Cybulski C, Lubinski J, Thibodeau SN, Schaid DJ, Sorensen KD, Batra J, Clements JA, Chambers S, Aitken J, Gardiner RA, Maier C, Vogel W, Dörk T, Brenner H, Habuchi T, Ingles S, John EM, Dickinson JL, Cannon-Albright L, Teixeira MR, Kaneva R, Zhang HW, Lu YJ, Park JY, Cooney KA, Muir KR, Leongamornlert DA, Saunders E, Tymrakiewicz M, Mahmud N, Guy M, Govindasami K, O'Brien LT, Wilkinson RA, Hall AL, Sawyer EJ, Dadaev T, Morrison J, Dearnaley DP, Horwich A, Huddart RA, Khoo VS, Parker CC, Van As N, Woodhouse CJ, Thompson A, Dudderidge T, Ogden C, Cooper CS, Lophatonanon A, Southey MC, Hopper JL, English D, Virtamo J, Le Marchand L, Campa D, Kaaks R, Lindstrom S, Diver WR, Gapstur S, Yeager M, Cox A, Stern MC, Corral R, Aly M, Isaacs W, Adolfsson J, Xu J, Zheng SL, Wahlfors T, Taari K, Kujala P, Klarskov P, Nordestgaard BG, Røder MA, Frikke-Schmidt R, Bojesen SE, FitzGerald LM, Kolb S, Kwon EM, Karyadi DM, Orntoft TF, Borre M, Rinckleb A, Luedeke M, Herkommer K, Meyer A, Serth J, Marthick JR, Patterson B, Wokolorczyk D, Spurdle A, Lose F, McDonnell SK, Joshi AD, Shahabi A, Pinto P, Santos J, Ray A, Sellers TA, Lin HY, Stephenson RA, Teerlink C, Muller H, Rothenbacher D, Tsuchiya N, Narita S, Cao GW, Slavov C, Mitev V, Chanock S, Gronberg H, Haiman CA, Kraft P, Easton DF, Eeles RA. A meta-analysis of genome-wide association studies to identify prostate cancer susceptibility loci associated with aggressive and non-aggressive disease. Hum Mol Genet 2013; 22:408-15. [PMID: 23065704 PMCID: PMC3526158 DOI: 10.1093/hmg/dds425] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 10/04/2012] [Indexed: 01/14/2023] Open
Abstract
Genome-wide association studies (GWAS) have identified multiple common genetic variants associated with an increased risk of prostate cancer (PrCa), but these explain less than one-third of the heritability. To identify further susceptibility alleles, we conducted a meta-analysis of four GWAS including 5953 cases of aggressive PrCa and 11 463 controls (men without PrCa). We computed association tests for approximately 2.6 million SNPs and followed up the most significant SNPs by genotyping 49 121 samples in 29 studies through the international PRACTICAL and BPC3 consortia. We not only confirmed the association of a PrCa susceptibility locus, rs11672691 on chromosome 19, but also showed an association with aggressive PrCa [odds ratio = 1.12 (95% confidence interval 1.03-1.21), P = 1.4 × 10(-8)]. This report describes a genetic variant which is associated with aggressive PrCa, which is a type of PrCa associated with a poorer prognosis.
Collapse
Affiliation(s)
- Ali Amin Al Olama
- Strangeways Laboratory, Centre for Cancer Genetic Epidemiology, Worts Causeway, Cambridge CB1 8RN, UK
| | - Zsofia Kote-Jarai
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Fredrick R. Schumacher
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, CA, USA
| | - Fredrik Wiklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm SE-171 77, Sweden
| | - Sonja I. Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Core Genotyping Facility, SAIC-Frederick, Inc., National Cancer Institute, NIH, Gaithersburg, MD, USA
| | - Sara Benlloch
- Strangeways Laboratory, Centre for Cancer Genetic Epidemiology, Worts Causeway, Cambridge CB1 8RN, UK
| | - Graham G. Giles
- Cancer Epidemiology Centre, Cancer Council Victoria, 1 Rathdowne Street, Carlton, VIC 3053, Australia
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, The University of Melbourne, 723 Swanston Street, Carlton, VIC 3053, Australia
| | - Gianluca Severi
- Cancer Epidemiology Centre, Cancer Council Victoria, 1 Rathdowne Street, Carlton, VIC 3053, Australia
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, The University of Melbourne, 723 Swanston Street, Carlton, VIC 3053, Australia
| | - David E. Neal
- Surgical Oncology (Uro-Oncology: S4), Addenbrooke's Hospital, University of Cambridge, Box 279, Hills Road, Cambridge, UK
- Li Ka Shing Centre, Cancer Research UK Cambridge Research Institute, Cambridge CB2 2QQ, UK
| | - Freddie C. Hamdy
- Nuffield Department of Surgery and
- Faculty of Medical Science, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Jenny L. Donovan
- School of Social and Community Medicine, University of Bristol, Canynge Hall, 39 Whatley Road, Bristol BS8 2PS, UK
| | - David J. Hunter
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology and
| | - Brian E. Henderson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, CA, USA
| | - Michael J. Thun
- Epidemiology Research Program, American Cancer Society, Atlanta, GA 30303, USA
| | - Michael Gaziano
- Massachusetts Veterans Epidemiology and Research Information Center (MAVERIC) and Geriatric Research, Education, and Clinical Center (GRECC), Boston Veterans Affairs Healthcare System, Boston, MA 02114, USA
- Division of Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA
| | | | - Afshan Siddiq
- Department of Genomics of Common Disease, School of Public Health, Imperial College, London SW7 2AZ, UK
| | - Ruth C. Travis
- Cancer Epidemiology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - David G. Cox
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
- Lyon Cancer Research Center, INSERM U1052, Lyon, France
| | | | - Elio Riboli
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
| | - Timothy J. Key
- Cancer Epidemiology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Gerald Andriole
- Division of Urologic Surgery, Washington University School of Medicine, St Louis, MO, USA
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Richard B. Hayes
- Division of Epidemiology, Department of Environmental Medicine, NYU Langone Medical Centre, NYU Cancer Institute, New York, NY 10016, USA
| | - Johanna Schleutker
- Institute of Biomedical Technology/BioMediTech, University of Tampere and
- Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland
| | - Anssi Auvinen
- Department of Epidemiology, School of Health Sciences and
| | - Teuvo L.J. Tammela
- Department of Urology, Tampere University Hospital and Medical School, University of Tampere, Tampere, Finland
| | | | - Janet L. Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Centre, Seattle, WA, USA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | - Elaine A. Ostrander
- National Human Genome Research Institute, National Institutes of Health, 50 South Drive, Room 5351, Bethesda, MD, USA
| | - Cezary Cybulski
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - Jan Lubinski
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | | | | | | | - Jyotsna Batra
- Australian Prostate Cancer Research Centre-Qld, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Judith A. Clements
- Australian Prostate Cancer Research Centre-Qld, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Suzanne Chambers
- Griffith Health Institute, Griffith University, Gold Coast, QLD, Australia
- Viertel Centre for Research in Cancer Control, Cancer Council Queensland, Brisbane, QLD, Australia
- Centre for Clinical Research, University of Queensland, Brisbane, QLD, Australia
| | - Joanne Aitken
- Viertel Centre for Research in Cancer Control, Cancer Council Queensland, Brisbane, QLD, Australia
| | - Robert A. Gardiner
- Centre for Clinical Research, University of Queensland, Brisbane, QLD, Australia
| | - Christiane Maier
- Department of Urology and
- Institute of Human Genetics, University Hospital Ulm, Ulm, Germany
| | - Walther Vogel
- Institute of Human Genetics, University Hospital Ulm, Ulm, Germany
| | - Thilo Dörk
- Hannover Medical School, Hannover, Germany
| | | | - Tomonori Habuchi
- Department of Urology,Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | - Sue Ingles
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, CA, USA
| | - Esther M. John
- Cancer Prevention Institute of California, Fremont, CA, USA
- Division of Epidemiology, Department of Health Research and Policy and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Joanne L. Dickinson
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, TAS, Australia
| | - Lisa Cannon-Albright
- Division of Genetic Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
- George E. Wahlen Department of Veterans Affairs Medical Centre, Salt Lake City, UT, USA
| | - Manuel R. Teixeira
- Department of Genetics, Portuguese Oncology Institute and Biomedical Sciences Institute (ICBAS), Porto University, Porto, Portugal
| | - Radka Kaneva
- Molecular Medicine Centre, Department of Medical Chemistry and Biochemistry, Medical University of Sofia, 2 Zdrave St, Sofia 1431, Bulgaria
| | - Hong-Wei Zhang
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | - Yong-Jie Lu
- Centre for Molecular Oncology and Imaging, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK
| | - Jong Y. Park
- Division of Cancer Prevention and Control, H. Lee Moffitt Cancer Centre, 12902 Magnolia Drive, Tampa, FL, USA
| | - Kathleen A. Cooney
- Department of Internal Medicine and
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | | | - Edward Saunders
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | | | - Nadiya Mahmud
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Michelle Guy
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Koveela Govindasami
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Lynne T. O'Brien
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | | | - Amanda L. Hall
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Emma J. Sawyer
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Tokhir Dadaev
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Jonathan Morrison
- Strangeways Laboratory, Centre for Cancer Genetic Epidemiology, Worts Causeway, Cambridge CB1 8RN, UK
| | - David P. Dearnaley
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| | - Alan Horwich
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| | - Robert A. Huddart
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| | - Vincent S. Khoo
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| | - Christopher C. Parker
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| | - Nicholas Van As
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| | | | - Alan Thompson
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| | - Tim Dudderidge
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| | - Chris Ogden
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| | - Colin S. Cooper
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | | | - Melissa C. Southey
- Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Grattan street, Parkville, VIC, Australia
| | - John L. Hopper
- Cancer Epidemiology Centre, Cancer Council Victoria, 1 Rathdowne Street, Carlton, VIC, Australia
| | - Dallas English
- Cancer Epidemiology Centre, Cancer Council Victoria, 1 Rathdowne Street, Carlton, VIC, Australia
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, The University of Melbourne, 723 Swanston Street, Carlton, VIC, Australia
| | - Jarmo Virtamo
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Centre, Department of Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Daniele Campa
- Lyon Cancer Research Center, INSERM U1052, Lyon, France
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Sara Lindstrom
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology and
| | - W. Ryan Diver
- Epidemiology Research Program, American Cancer Society, Atlanta, GA 30303, USA
| | - Susan Gapstur
- Epidemiology Research Program, American Cancer Society, Atlanta, GA 30303, USA
| | - Meredith Yeager
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Core Genotyping Facility, SAIC-Frederick, Inc., National Cancer Institute, NIH, Gaithersburg, MD, USA
| | - Angela Cox
- Department of Oncology, University of Sheffield, Sheffield, UK
| | - Mariana C. Stern
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, CA, USA
| | - Roman Corral
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, CA, USA
| | - Markus Aly
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm SE-171 77, Sweden
- Division of Urology, Department of Clinical Sciences, Danderyd Hospital and
| | - William Isaacs
- School of Medicine, Johns Hopkins University, 115 Marburg Building, 600 North Wolfe Street, Baltimore, MD 21205, USA
| | - Jan Adolfsson
- Oncological Centre, CLINTEC, Karolinska Institute, Stockholm, Sweden
| | - Jianfeng Xu
- Center for Cancer Genomics, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - S. Lilly Zheng
- Center for Cancer Genomics, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Tiina Wahlfors
- Department of Urology, Tampere University Hospital and Medical School, University of Tampere, Tampere, Finland
| | - Kimmo Taari
- Department of Urology, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
| | - Paula Kujala
- Department of Pathology, Centre for Laboratory Medicine, Tampere University Hospital, Tampere, Finland
| | - Peter Klarskov
- Department of Urology, Herlev Hospital, Copenhagen University Hospital, Herlev Ringvej 75, Herlev DK-2730, Denmark
| | | | | | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, Copenhagen DK-2100, Denmark
| | | | - Liesel M. FitzGerald
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Centre, Seattle, WA, USA
| | - Suzanne Kolb
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Centre, Seattle, WA, USA
| | - Erika M. Kwon
- National Human Genome Research Institute, National Institutes of Health, 50 South Drive, Room 5351, Bethesda, MD, USA
| | - Danielle M. Karyadi
- National Human Genome Research Institute, National Institutes of Health, 50 South Drive, Room 5351, Bethesda, MD, USA
| | | | - Michael Borre
- Department of Urology, Aarhus University Hospital, Skejby, Denmark
| | | | - Manuel Luedeke
- Institute of Human Genetics, University Hospital Ulm, Ulm, Germany
| | - Kathleen Herkommer
- Department of Urology, Rechts der Isar Medical Centre, Technical University of Munich, Munich, Germany
| | | | | | - James R. Marthick
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, TAS, Australia
| | - Briony Patterson
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, TAS, Australia
| | - Dominika Wokolorczyk
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | | | - Felicity Lose
- Molecular Cancer Epidemiology Laboratory, Queensland Institute of Medical Research, Brisbane, QLD, Australia
| | | | - Amit D. Joshi
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, CA, USA
| | - Ahva Shahabi
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, CA, USA
| | - Pedro Pinto
- Department of Genetics, Portuguese Oncology Institute and Biomedical Sciences Institute (ICBAS), Porto University, Porto, Portugal
| | - Joana Santos
- Department of Genetics, Portuguese Oncology Institute and Biomedical Sciences Institute (ICBAS), Porto University, Porto, Portugal
| | - Ana Ray
- Department of Internal Medicine and
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Thomas A. Sellers
- Division of Cancer Prevention and Control, H. Lee Moffitt Cancer Centre, 12902 Magnolia Drive, Tampa, FL, USA
| | - Hui-Yi Lin
- Division of Cancer Prevention and Control, H. Lee Moffitt Cancer Centre, 12902 Magnolia Drive, Tampa, FL, USA
| | | | - Craig Teerlink
- Division of Genetic Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Heiko Muller
- Division of Clinical Epidemiology and Aging Research and
| | | | - Norihiko Tsuchiya
- Department of Urology,Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | - Shintaro Narita
- Department of Urology,Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | - Guang-Wen Cao
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | - Chavdar Slavov
- Department of Urology and Alexandrovska University Hospital,Medical University of Sofia, Sofia, Bulgaria
| | - Vanio Mitev
- Molecular Medicine Centre, Department of Medical Chemistry and Biochemistry, Medical University of Sofia, 2 Zdrave St, Sofia 1431, Bulgaria
| | | | | | | | | | - Stephen Chanock
- Core Genotyping Facility, SAIC-Frederick, Inc., National Cancer Institute, NIH, Gaithersburg, MD, USA
| | - Henrik Gronberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm SE-171 77, Sweden
| | - Christopher A. Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, CA, USA
| | - Peter Kraft
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology and
| | - Douglas F. Easton
- Strangeways Laboratory, Centre for Cancer Genetic Epidemiology, Worts Causeway, Cambridge CB1 8RN, UK
| | - Rosalind A. Eeles
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| |
Collapse
|
48
|
Diversity of human clock genotypes and consequences. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 119:51-81. [PMID: 23899594 DOI: 10.1016/b978-0-12-396971-2.00003-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The molecular clock consists of a number of genes that form transcriptional and posttranscriptional feedback loops, which function together to generate circadian oscillations that give rise to circadian rhythms of our behavioral and physiological processes. Genetic variations in these clock genes have been shown to be associated with phenotypic effects in a repertoire of biological processes, such as diurnal preference, sleep, metabolism, mood regulation, addiction, and fertility. Consistently, rodent models carrying mutations in clock genes also demonstrate similar phenotypes. Taken together, these studies suggest that human clock-gene variants contribute to the phenotypic differences observed in various behavioral and physiological processes, although to validate this requires further characterization of the molecular consequences of these polymorphisms. Investigating the diversity of human genotypes and the phenotypic effects of these genetic variations shall advance our understanding of the function of the circadian clock and how we can employ the clock to improve our overall health.
Collapse
|
49
|
Sheikh H, Abdulghani J, Ali S, Sinha R, Lipton A. Impact of Genetic Targets on Prostate Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 779:359-83. [DOI: 10.1007/978-1-4614-6176-0_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
50
|
Abstract
Screening for prostate cancer is a controversial topic within the field of urology. The US Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial did not demonstrate any difference in prostate-cancer-related mortality rates between men screened annually rather than on an 'opportunistic' basis. However, in the world's largest trial to date--the European Randomised Study of Screening for Prostate Cancer--screening every 2-4 years was associated with a 21% reduction in prostate-cancer-related mortality rate after 11 years. Citing the uncertain ratio between potential harm and potential benefit, the US Preventive Services Task Force recently recommended against serum PSA screening. Although this ratio has yet to be elucidated, PSA testing--and early tumour detection--is undoubtedly beneficial for some individuals. Instead of adopting a 'one size fits all' approach, physicians are likely to perform personalized risk assessment to minimize the risk of negative consequences, such as anxiety, unnecessary testing and biopsies, overdiagnosis, and overtreatment. The PSA test needs to be combined with other predictive factors or be used in a more thoughtful way to identify men at risk of symptomatic or life-threatening cancer, without overdiagnosing indolent disease. A risk-adapted approach is needed, whereby PSA testing is tailored to individual risk.
Collapse
|