1
|
Schroeder C, Campilan B, Leary OP, Arditi J, Michles MJ, De La Garza Ramos R, Akinduro OO, Gokaslan ZL, Martinez Moreno M, Sullivan PLZ. Therapeutic Opportunities for Biomarkers in Metastatic Spine Tumors. Cancers (Basel) 2024; 16:3152. [PMID: 39335124 PMCID: PMC11430692 DOI: 10.3390/cancers16183152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/01/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
For many spine surgeons, patients with metastatic cancer are often present in an emergent situation with rapidly progressive neurological dysfunction. Since the Patchell trial, scoring systems such as NOMS and SINS have emerged to guide the extent of surgical excision and fusion in the context of chemotherapy and radiation therapy. Yet, while multidisciplinary decision-making is the gold standard of cancer care, in the middle of the night, when a patient needs spinal surgery, the wealth of chemotherapy data, clinical trials, and other medical advances can feel overwhelming. The goal of this review is to provide an overview of the relevant molecular biomarkers and therapies driving patient survival in lung, breast, prostate, and renal cell cancer. We highlight the molecular differences between primary tumors (i.e., the patient's original lung cancer) and the subsequent spinal metastasis. This distinction is crucial, as there are limited data investigating how metastases respond to their primary tumor's targeted molecular therapies. Integrating information from primary and metastatic markers allows for a more comprehensive and personalized approach to cancer treatment.
Collapse
Affiliation(s)
- Christian Schroeder
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Beatrice Campilan
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Owen P Leary
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Jonathan Arditi
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Madison J Michles
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Rafael De La Garza Ramos
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Oluwaseun O Akinduro
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Ziya L Gokaslan
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Margot Martinez Moreno
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | | |
Collapse
|
2
|
Purkayastha A, Roy A, Bharadaj S, Bharadaj SK, Chakraborty S. Turning off a few overexpressed genes in prostate cancer with microRNAs using a 7mer-seed match model. J Cancer Res Clin Oncol 2023; 149:10335-10364. [PMID: 37273107 DOI: 10.1007/s00432-023-04910-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 05/20/2023] [Indexed: 06/06/2023]
Abstract
PURPOSE This research aims to identify the miRNAs that could target the genes overexpressed in prostate cancer so that miRNA-based therapeutics could be developed. METHODS A 7mer-m8 model of microRNA targeting was utilized in order to analyse the relationship between microRNAs and overexpressed genes. The efficiency of miRNA binding was investigated using various parameters namely free energy (AMFE), GC and GC3 content, translation efficiency, cosine similarity metric, mRNA stability, free energy of RNA duplex, and base compositional difference. BLAST2GO software was used to elucidate the functional roles of the genes overexpressed in prostate cancer. RESULTS The current research reveals that the coding sequences of the genes were found targeted with multiple miRNAs. For instance, the HPN gene was targeted by the microRNA miR-4279 at two distinct sites i.e. 263-278 and 746-761 in the coding sequence. In the present study, it was observed that the target region of the genes exhibited a comparatively high GC and GC3 contents in comparison to the flanking regions. A low translational rate and weak relationship between RSCU and tRNA were obtained which may be due to the absence of optimal codons. CONCLUSION In this study, we have uncovered the human miRNAs that have potential for binding to the coding sequences of 14 most overexpressed genes in prostate cancer and thereby could silence those genes.
Collapse
Affiliation(s)
- Arpita Purkayastha
- Department of Biotechnology, Assam University, Silchar, Assam, 788011, India
| | - Aparajita Roy
- Department of Biotechnology, Assam University, Silchar, Assam, 788011, India
| | - Stella Bharadaj
- Silchar Medical College and Hospital, Silchar, Assam, 788014, India
| | | | - Supriyo Chakraborty
- Department of Biotechnology, Assam University, Silchar, Assam, 788011, India.
| |
Collapse
|
3
|
Stamis SA, Heath EI, Lucas S, Boerner J, Slusher LB. Alcohol dehydrogenase expression patterns in normal prostate, benign prostatic hyperplasia, and prostatic adenocarcinoma in African American and Caucasian men. Prostate 2022; 82:666-675. [PMID: 35133686 DOI: 10.1002/pros.24310] [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: 03/15/2021] [Revised: 01/09/2022] [Accepted: 01/14/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND In situ metabolism of ethanol by alcohol dehydrogenases (ADHs) contributes to oxidative damage of cells and DNA and has been linked to carcinogenesis in numerous epithelial tissues. The goal of this study was to determine expression patterns of ADH1 and ADH7 isozymes in normal, hyperplastic (benign prostatic hyperplasia [BPH]) and neoplastic (prostate cancer [PCa]) prostate. Furthermore, racial differences in ADH expression between African Americans and Caucasians were investigated. METHODS ADH expression patterns were characterized by density analysis of ADH immunohistochemistry (n = 21) and real-time RT-PCR of total RNAs by laser-capture microdissection (n = 10) and whole tissue formalin-fixed paraffin embedded prostate biopsies (n = 63). RESULTS ADH protein is found in normal prostate and is primarily associated with glandular epithelium. Transcripts of ADH1B are suppressed in PCa compared to BPH (p = 0.0095). Racial differences in ADH7 transcripts exist between African American and Caucasian men. A total of 57.6% of biopsies from African American prostates have detectable ADH7 messenger RNA (mRNA) transcripts compared to the 13.3% of Caucasian prostate biopsies with detectable transcripts (p = 0.0005). This increased frequency of detection contributes to higher mean ADH7 mRNA transcript levels in African Americans (p = 0.001). CONCLUSIONS To our knowledge this study is the first to report downregulation of ADH1B in neoplastic prostate at the transcriptional level, suggesting protective regulatory functions. ADH7 transcripts were not detectable in all samples and was found in higher frequency and amount in our African American samples. Racial differences in ADH7 within the prostate is a novel finding and should be investigated further.
Collapse
Affiliation(s)
- Sarah A Stamis
- Department of Biology, West Chester University of Pennsylvania, West Chester, Pennsylvania, USA
| | - Elisabeth I Heath
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Steven Lucas
- Department of Urology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Julie Boerner
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Leslie B Slusher
- Department of Biology, West Chester University of Pennsylvania, West Chester, Pennsylvania, USA
| |
Collapse
|
4
|
Pavel AG, Stambouli D, Gener I, Preda A, Anton G, Baston C. Genetic variant located on chromosome 17p12 contributes to prostate cancer onset and biochemical recurrence. Sci Rep 2022; 12:4546. [PMID: 35296725 PMCID: PMC8927158 DOI: 10.1038/s41598-022-08472-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 03/07/2022] [Indexed: 12/24/2022] Open
Abstract
The genetic contribution to prostate cancer (PC) onset and clinical heterogeneity has an important impact on the disease stratification accuracy. Despite the fact that radical prostatectomy (RP) is an effective treatment for localized PC, a considerable number of individuals develop biochemical recurrence (BCR) following surgery. In the present study, we decided to investigate the significance of genetic variability in a homogeneous group of Romanian men and to determine if genotyping could provide information regarding the possible implications of rs4054823 susceptibility loci in PC progression and outcome. A total of 78 samples from both PC and benign prostatic hyperplasia (BPH) patients were genotyped. The genotype frequencies were examined to see if there was a link between the 17p12 SNP and PC disease. When compared to the BPH group, the PC group had a significantly higher frequency of the T risk variant (P = 0.0056) and TT genotype (P = 0.0164). Subsequent analysis revealed that the TT genotype had a significantly higher frequency among younger PC patients based on their age at diagnosis and that it was related with a greater probability of BCR (P = 0.02). According to our findings, the TT genotype appears to be a risk factor for early-onset PC and a potential predictor for BCR after RP.
Collapse
Affiliation(s)
- Anca Gabriela Pavel
- Molecular Genetics Department, Cytogenomic Medical Laboratory, Bucharest, Romania. .,The Romania Academy, "Stefan S. Nicolau" Institute of Virology, Bucharest, Romania.
| | - Danae Stambouli
- Molecular Genetics Department, Cytogenomic Medical Laboratory, Bucharest, Romania
| | - Ismail Gener
- Department of Nephrology, Urology, Immunology and Immunology of Transplant, Dermatology, Allergology, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.,Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| | - Adrian Preda
- Center of Urological Surgery, Dialysis and Renal Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Gabriela Anton
- The Romania Academy, "Stefan S. Nicolau" Institute of Virology, Bucharest, Romania
| | - Catalin Baston
- Department of Nephrology, Urology, Immunology and Immunology of Transplant, Dermatology, Allergology, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.,Center of Urological Surgery, Dialysis and Renal Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| |
Collapse
|
5
|
Hamid ARAH, Luna-Velez MV, Dudek AM, Jansen CFJ, Smit F, Aalders TW, Verhaegh GW, Schaafsma E, Sedelaar JPM, Schalken JA. Molecular Phenotyping of AR Signaling for Predicting Targeted Therapy in Castration Resistant Prostate Cancer. Front Oncol 2021; 11:721659. [PMID: 34490120 PMCID: PMC8417043 DOI: 10.3389/fonc.2021.721659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 07/31/2021] [Indexed: 11/13/2022] Open
Abstract
Castration-resistant prostate cancer (CRPC) is defined by resistance of the tumor to androgen deprivation therapy (ADT). Several molecular changes, particularly in the AR signaling cascade, have been described that may explain ADT resistance. The variety of changes may also explain why the response to novel therapies varies between patients. Testing the specific molecular changes may be a major step towards personalized treatment of CRPC patients. The aim of our study was to evaluate the molecular changes in the AR signaling cascade in CRPC patients. We have developed and validated several methods which are easy to use, and require little tissue material, for exploring AR signaling pathway changes simultaneously. We found that the AR signaling pathway is still active in the majority of our CRPC patients, due to molecular changes in AR signaling components. There was heterogeneity in the molecular changes observed, but we could classify the patients into 4 major subgroups which are: AR mutation, AR amplification, active intratumoral steroidogenesis, and combination of AR amplification and active intratumoral steroidogenesis. We suggest characterizing the AR signaling pathway in CRPC patients before beginning any new treatment, and a recent fresh tissue sample from the prostate or a metastatic site should be obtained for the purpose of this characterization.
Collapse
Affiliation(s)
- Agus Rizal A H Hamid
- Department of Urology, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Urology, Ciptomangunkusumo Hospital, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Maria V Luna-Velez
- Department of Urology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Aleksandra M Dudek
- Department of Urology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | | | - Tilly W Aalders
- Department of Urology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Gerald W Verhaegh
- Department of Urology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ewout Schaafsma
- Department of Pathology, Radboud University Medical Center, Nijmegen, Netherlands
| | - John P M Sedelaar
- Department of Urology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jack A Schalken
- Department of Urology, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
6
|
Santos NJ, Barquilha CN, Barbosa IC, Macedo RT, Lima FO, Justulin LA, Barbosa GO, Carvalho HF, Felisbino SL. Syndecan Family Gene and Protein Expression and Their Prognostic Values for Prostate Cancer. Int J Mol Sci 2021; 22:ijms22168669. [PMID: 34445387 PMCID: PMC8395474 DOI: 10.3390/ijms22168669] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is the leading cause of cancer-associated mortality in men, and new biomarkers are still needed. The expression pattern and protein tissue localization of proteoglycans of the syndecan family (SDC 1-4) and syntenin-1 (SDCBP) were determined in normal and prostatic tumor tissue from two genetically engineered mouse models and human prostate tumors. Studies were validated using SDC 1-4 and SDCBP mRNA levels and patient survival data from The Cancer Genome Atlas and CamCAP databases. RNAseq showed increased expression of Sdc1 in Pb-Cre4/Ptenf/f mouse Pca and upregulation of Sdc3 expression and downregulation of Sdc2 and Sdc4 when compared to the normal prostatic tissue in Pb-Cre4/Trp53f/f-;Rb1f/f mouse tumors. These changes were confirmed by immunohistochemistry. In human PCa, SDC 1-4 and SDCBP immunostaining showed variable localization. Furthermore, Kaplan-Meier analysis showed that patients expressing SDC3 had shorter prostate-specific survival than those without SDC3 expression (log-rank test, p = 0.0047). Analysis of the MSKCC-derived expression showed that SDC1 and SDC3 overexpression is predictive of decreased biochemical recurrence-free survival (p = 0.0099 and p = 0.045, respectively), and SDC4 overexpression is predictive of increased biochemical recurrence-free survival (p = 0.035). SDC4 overexpression was associated with a better prognosis, while SDC1 and SDC3 were associated with more aggressive tumors and a worse prognosis.
Collapse
Affiliation(s)
- Nilton José Santos
- Department of Structural and Functional BIology, Institute of Bioscience of Botucatu (IBB), São Paulo State University, Botucatu 18618-689, SP, Brazil; (N.J.S.); (C.N.B.); (I.C.B.); (L.A.J.)
- Department of Structural and Functional Biology, Institute of Biology (IB), UNICAMP—State University of Campinas, Campinas 13083-970, SP, Brazil; (G.O.B.); (H.F.C.)
| | - Caroline Nascimento Barquilha
- Department of Structural and Functional BIology, Institute of Bioscience of Botucatu (IBB), São Paulo State University, Botucatu 18618-689, SP, Brazil; (N.J.S.); (C.N.B.); (I.C.B.); (L.A.J.)
- Department of Structural and Functional Biology, Institute of Biology (IB), UNICAMP—State University of Campinas, Campinas 13083-970, SP, Brazil; (G.O.B.); (H.F.C.)
| | - Isabela Correa Barbosa
- Department of Structural and Functional BIology, Institute of Bioscience of Botucatu (IBB), São Paulo State University, Botucatu 18618-689, SP, Brazil; (N.J.S.); (C.N.B.); (I.C.B.); (L.A.J.)
- Department of Structural and Functional Biology, Institute of Biology (IB), UNICAMP—State University of Campinas, Campinas 13083-970, SP, Brazil; (G.O.B.); (H.F.C.)
| | - Rodrigo Tavares Macedo
- Botucatu School of Medicine (FMB), São Paulo State University, Botucatu 01049-010, SP, Brazil; (R.T.M.); (F.O.L.)
| | - Flávio Oliveira Lima
- Botucatu School of Medicine (FMB), São Paulo State University, Botucatu 01049-010, SP, Brazil; (R.T.M.); (F.O.L.)
| | - Luis Antônio Justulin
- Department of Structural and Functional BIology, Institute of Bioscience of Botucatu (IBB), São Paulo State University, Botucatu 18618-689, SP, Brazil; (N.J.S.); (C.N.B.); (I.C.B.); (L.A.J.)
| | - Guilherme Oliveira Barbosa
- Department of Structural and Functional Biology, Institute of Biology (IB), UNICAMP—State University of Campinas, Campinas 13083-970, SP, Brazil; (G.O.B.); (H.F.C.)
| | - Hernandes F. Carvalho
- Department of Structural and Functional Biology, Institute of Biology (IB), UNICAMP—State University of Campinas, Campinas 13083-970, SP, Brazil; (G.O.B.); (H.F.C.)
| | - Sérgio Luis Felisbino
- Department of Structural and Functional BIology, Institute of Bioscience of Botucatu (IBB), São Paulo State University, Botucatu 18618-689, SP, Brazil; (N.J.S.); (C.N.B.); (I.C.B.); (L.A.J.)
- Correspondence:
| |
Collapse
|
7
|
Zhang L, Li Y, Wang X, Ping Y, Wang D, Cao Y, Dai Y, Liu W, Tao Z. Five-gene signature associating with Gleason score serve as novel biomarkers for identifying early recurring events and contributing to early diagnosis for Prostate Adenocarcinoma. J Cancer 2021; 12:3626-3647. [PMID: 33995639 PMCID: PMC8120165 DOI: 10.7150/jca.52170] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Compared to non-recurrent type, recurrent prostate adenocarcinoma (PCa) is highly fatal, and significantly shortens the survival time of affected patients. Early and accurate laboratory diagnosis is particularly important in identifying patients at high risk of recurrence, necessary for additional systemic intervention. We aimed to develop efficient and accurate diagnostic and prognostic biomarkers for new PCa following radical therapy. Methods: We identified differentially expressed genes (DEGs) and clinicopathological data of PCa patients from Gene Expression Omnibus (GEO) datasets and The Cancer Genome Atlas (TCGA) repositories. We then uncovered the most relevant clinical traits and genes modules associated with PCa prognosis using the Weighted gene correlation network analysis (WGCNA). Univariate Cox regression analysis and multivariate Cox proportional hazards (Cox-PH) models were performed to identify candidate gene signatures related to Disease-Free Interval (DFI). Data for internal and external cohorts were utilized to test and validate the accuracy and clinical utility of the prognostic models. Results: We constructed and validated an accurate and reliable model for predicting the prognosis of PCa using 5 Gleason score-associated gene signatures (ZNF695, CENPA, TROAP, BIRC5 and KIF20A). The ROC and Kaplan-Meier analysis revealed the model was highly accurate in diagnosing and predicting the recurrence and metastases of PCa. The accuracy of the model was validated using the calibration curves based on internal TCGA cohort and external GEO cohort. Using the model, patients could be prognostically stratified in to various groups including TNM classification and Gleason score. Multivariate analysis revealed the model could independently predict the prognosis of PCa patients and its utility was superior to that of clinicopathological characteristics. In addition, we fund the expression of the 5 gene signatures strongly and positively correlated with tumor purity but negatively correlated with infiltration CD8+ T cells to the tumor microenvironment. Conclusions: A 5 gene signatures can accurately be used in the diagnosis and prediction of PCa prognosis. Thus this can guide the treatment and management prostate adenocarcinoma.
Collapse
Affiliation(s)
- Lingyu Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yu Li
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Anhui 233030, China
| | - Xuchu Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Ying Ping
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Danhua Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Ying Cao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yibei Dai
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Weiwei Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Zhihua Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| |
Collapse
|
8
|
Shiota M, Akamatsu S, Narita S, Terada N, Fujimoto N, Eto M. Genetic Polymorphisms and Pharmacotherapy for Prostate Cancer. JMA J 2021; 4:99-111. [PMID: 33997443 PMCID: PMC8119070 DOI: 10.31662/jmaj.2021-0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 01/15/2021] [Indexed: 11/17/2022] Open
Abstract
The therapeutic landscape of pharmacotherapy for prostate cancer has dramatically evolved, and multiple therapeutic options have become available for prostate cancer patients. Therefore, useful biomarkers to identify suitable candidates for treatment are required to maximize the efficacy of pharmacotherapy. Genetic polymorphisms such as single-nucleotide polymorphisms (SNPs) and tandem repeats have been shown to influence the therapeutic effects of pharmacotherapy for prostate cancer patients. For example, genetic polymorphisms in the genes involved in androgen receptor signaling are reported to be associated with the therapeutic outcome of androgen-deprivation therapy as well as androgen receptor-pathway inhibitors. In addition, SNPs in genes involved in drug metabolism and efflux pumps are associated with therapeutic effects of taxane chemotherapy. Thus, genetic polymorphisms such as SNPs are promising biomarkers to realize personalized medicine. Here, we overview the current findings on the influence of genetic polymorphisms on the outcome of pharmacotherapy for prostate cancer and discuss current issues as well as future visions in this field.
Collapse
Affiliation(s)
- Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shusuke Akamatsu
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shintaro Narita
- Department of Urology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Naoki Terada
- Department of Urology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Naohiro Fujimoto
- Department of Urology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Masatoshi Eto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
9
|
Abstract
Prostate cancer is the second leading cause of cancer-related deaths among male population worldwide, its incidence and lethality steadily increase. Nuclear enriched abundant transcript 1 (NEAT1) is a long non-coding RNA (ncRNA), located on chromatin 11. It has been found to function as an oncogene in different kinds of cancer. However, until now, the clinical significance of NEAT1 has not been investigated in prostate cancer.Paired tissue specimens of prostate cancer and matched normal prostate tissues were obtained from 130 patients with prostate cancer between 2014 and 2019 at The Fourth Affiliated Hospital Zhejiang University, School of Medicine. Group means were compared using the Student t test. Chi-Squared test was used for analyzing the correlation of the expression of NEAT1 with clinicopathologic features of prostate cancer patients. Survival data was analyzed using the Kaplan-Meier estimate and log-rank P was calculated. Cox regression model was used for univariate and multivariate analysis for factors related to overall survival.The expression of NEAT1 was increased significantly in prostate cancer tissues, compared with adjacent normal prostate tissues (P < .001). NEAT1 expression was significantly associated with TNM stage (P = .005), lymph nodes metastasis (P = .005), distant metastasis(P = .003), and Gleason score (P = .001). Overall survival rate was significantly lower for prostate cancer patients with a high expression level of NEAT1 than those with a low NEAT1a expression level (P = .048). In multivariate analysis, the results showed that the expression of NEAT1 was an independent prognostic factor for overall patient survival (HR: 2.111, CI: 1.735-10.295, P = .039).In the present study, NEAT1 is identified as an important lncRNA that may predict the prognosis of patients with prostate cancer.
Collapse
Affiliation(s)
- Junchao Bai
- Department of Urology, The Fourth Affiliated Hospital Zhejiang University, School of Medicine, Yiwu, Zhejiang, China
| | | |
Collapse
|
10
|
Sulfiredoxin as a Potential Therapeutic Target for Advanced and Metastatic Prostate Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2148562. [PMID: 32411320 PMCID: PMC7201699 DOI: 10.1155/2020/2148562] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/09/2019] [Indexed: 12/28/2022]
Abstract
The incidence of prostate cancer (PCa) is increasing, and it is currently the second most frequent cause of death by cancer in men. Despite advancements in cancer therapies, new therapeutic approaches are still needed for treatment-refractory advanced metastatic PCa. Cross-species analysis presents a robust strategy for the discovery of new potential therapeutic targets. This strategy involves the integration of genomic data from genetically engineered mouse models (GEMMs) and human PCa datasets. Considering the role of antioxidant pathways in tumor initiation and progression, we searched oxidative stress-related genes for a potential therapeutic target for PCa. First, we analyzed RNA-sequencing data from Pb-Cre4; Ptenf/f mice and discovered an increase in sulfiredoxin (Srxn1) mRNA expression in high-grade prostatic intraepithelial neoplasia (PIN), well-differentiated adenocarcinoma (medium-stage tumors), and poor-differentiated adenocarcinoma (advanced-stage prostate tumors). The increase of SRXN1 protein expression was confirmed by immunohistochemistry in mouse prostate tumor paraffin samples. Analyses of human databases and prostate tissue microarrays demonstrated that SRXN1 is overexpressed in a subset of high-grade prostate tumors and correlates with aggressive PCa with worse prognosis and decreased survival. Analyses in vitro showed that SRXN1 expression is also higher in most PCa cell lines compared to normal cell lines. Furthermore, siRNA-mediated downregulation of SRXN1 led to decreased viability of PCa cells LNCaP. In conclusion, we identified the antioxidant enzyme SRXN1 as a potential therapeutic target for PCa. Our results suggest that the use of specific SRXN1 inhibitors may be an effective strategy for the adjuvant treatment of castration-resistant PCa with SRXN1 overexpression.
Collapse
|
11
|
Aldaoud N, Hallak A, Abdo N, Al Bashir S, Marji N, Graboski-Bauer A. Interobserver Variability in the Diagnosis of High-Grade Prostatic Intraepithelial Neoplasia in a Tertiary Hospital in Northern Jordan. CLINICAL PATHOLOGY 2020; 13:2632010X19898472. [PMID: 31950103 PMCID: PMC6952849 DOI: 10.1177/2632010x19898472] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 12/06/2018] [Indexed: 11/16/2022]
Abstract
Prostate intraepithelial neoplasia is described as a precursor lesion to prostatic adenocarcinoma. High-grade prostate intraepithelial neoplasia (HGPIN) is classified as both grade 2 and 3 prostate intraepithelial neoplasia due to inconsistency between pathologists' findings. In our study, we assessed the interobserver variability in the diagnosis of HGPIN among genitourinary and nongenitourinary pathologists. All cases with prostate adenocarcinoma diagnosis on needle core biopsy, radical prostatectomy, and transurethral resection of prostate (TURP) between the years 2005 and 2014 were included. In total, 191 prostate cancer cases were included: 109 needle core biopsies, 45 radical prostatectomies, and 37 TURP. All were independently reviewed by 2 urologic pathologists for the presence of HGPIN. High-grade prostate intraepithelial neoplasia was diagnosed in 65 cases (34%), among which the lesion was recognized by the reporting pathologists in 36 (55%) of the cases and was missed in 29 (45%) of the cases with a κ coefficient of 0.53. There was a moderate interobserver agreement in the diagnosis of HGPIN. Consultation with genitourinary pathologist can improve HGPIN diagnosis.
Collapse
Affiliation(s)
- Najla Aldaoud
- Department of Pathology and Microbiology, Jordan University of Science and Technology, Irbid, Jordan.,Department of Pathology and Microbiology, King Abdullah University Hospital, Irbid, Jordan
| | - Amer Hallak
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Nour Abdo
- Department of Public Health, Jordan University of Science and Technology, Irbid, Jordan
| | - Samir Al Bashir
- Department of Pathology and Microbiology, Jordan University of Science and Technology, Irbid, Jordan
| | - Noor Marji
- Department of Pathology and Microbiology, Jordan University of Science and Technology, Irbid, Jordan.,Department of Pathology, University of Florida, Jacksonville, FL, USA
| | - Ashley Graboski-Bauer
- Department of Public Health Sciences, New Mexico State University, Las Cruces, NM, USA
| |
Collapse
|
12
|
Dong H, Hu J, Wang L, Qi M, Lu N, Tan X, Yang M, Bai X, Zhan X, Han B. SOX4 is activated by C-MYC in prostate cancer. Med Oncol 2019; 36:92. [PMID: 31560094 DOI: 10.1007/s12032-019-1317-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/12/2019] [Indexed: 11/30/2022]
Abstract
Although MYC proto-oncogene (C-MYC) amplification has been consistently reported to be a potential marker for prostate cancer (PCa) progression and prognosis, the clinicopathological and prognostic significance of C-MYC protein expression remains controversial. Overexpression of SOX4 has been shown to play important roles in multiple cancers including PCa. However, the link between these two critical genetic aberrations is unclear. In the current study, we showed that C-MYC was overexpressed in 16.2% (17/105) of Chinese patients with localized PCa. Overexpression of C-MYC was significantly associated with high Gleason scores (P = 0.012) and high Ki67 labeling index (P = 0.005). C-MYC overexpression was correlated with cancer-related mortality and suggested to be an unfavorable prognostic factor in Chinese PCa patients (P = 0.018). Overexpression of C-MYC is associated with SOX4 overexpression in PCa tissues. Notably, SOX4 is a direct target gene of C-MYC; C-MYC activates SOX4 expression via binding to its promoter. In addition, Co-IP analysis demonstrated a physical interaction between C-MYC and SOX4 protein in PCa cells. Clinically, C-MYC+/SOX4+ characterized poor prognosis in a subset of PCa patients. In total, C-MYC overexpression may contribute to PCa progression by activating SOX4. Our findings highlight an important role of C-MYC/SOX4 in PCa progression in a subset of PCa patients.
Collapse
Affiliation(s)
- Hongyan Dong
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
- Department of Pathology, Linyi People's Hospital, Linyi, China
| | - Jing Hu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Lin Wang
- Research Center for Medical Biotechnology, Shandong Academy of Medical Sciences, Jinan, China
| | - Mei Qi
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Ning Lu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Xiao Tan
- Department of Pathology, Linyi People's Hospital, Linyi, China
| | - Muyi Yang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Xinnuo Bai
- Department of Human Biology, University of Toronto, Toronto, M5S3J6, Canada
| | - Xuemei Zhan
- Department of Pathology, Linyi People's Hospital, Linyi, China
| | - Bo Han
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China.
- Department of Pathology, Shandong University Qilu Hospital, Jinan, China.
| |
Collapse
|
13
|
Aldaoud N, Graboski-Bauer A, Abdo N, Al Bashir S, Oweis AO, Ebwaini H, Hasen Y, Alazab R, Trpkov K. ERG expression in prostate cancer biopsies with and without high-grade prostatic intraepithelial neoplasia: a study in Jordanian Arab patients. Res Rep Urol 2019; 11:149-155. [PMID: 31192172 PMCID: PMC6535407 DOI: 10.2147/rru.s207843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 04/15/2019] [Indexed: 11/23/2022] Open
Abstract
Background: High-grade prostatic intraepithelial neoplasia (HGPIN) is the most likely precancerous lesion for prostatic adenocarcinoma (PCa). Recent molecular studies have shown that HGPIN can harbor TMPRSS2-ERG fusion, a genetic marker also associated with PCa, which may provide an additional risk stratification tool for HGPIN, especially when present as an isolated lesion. Our aim was to assess the frequency of HGPIN and ERG expression in a cohort of prostatic needle core biopsies from Jordanian-Arab patients with PCa. Materials and methods: We studied 109 needle core biopsies from patients with PCa. Clinical data, including age and preoperative prostate specific antigen (PSA) level, were obtained from patients’ medical records. Results: HGPIN was present in 31 (28.4 %) of the 109 cases. Of the HGPIN cases, 13 (41.9%) expressed ERG immunostain. ERG expression in HGPIN was independent of patient age at presentation (P=0.4), pre-operative PSA (P=0.9), and the grade, using the novel Grade Groups (P=0.5). Conclusion: The frequency of HGPIN in our cohort appears similar to the one found in the Western patient populations and demonstrates a comparable frequency of ERG expression in these lesions.
Collapse
Affiliation(s)
- Najla Aldaoud
- Department of Pathology and Microbiology, King Abdullah University Hospital, Irbid, Jordan.,Jordan University of Science and Technology, Irbid, Jordan
| | | | - Nour Abdo
- Department of Public Health, Jordan University of Science and Technology, Irbid, Jordan
| | - Samir Al Bashir
- Department of Pathology and Microbiology, King Abdullah University Hospital, Irbid, Jordan.,Jordan University of Science and Technology, Irbid, Jordan
| | - Ashraf O Oweis
- Division of Nephrology, Department of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Hanadi Ebwaini
- Department of Pathology and Microbiology, King Abdullah University Hospital, Irbid, Jordan
| | - Yousef Hasen
- Department of Pathology and Microbiology, King Abdullah University Hospital, Irbid, Jordan.,Jordan University of Science and Technology, Irbid, Jordan.,Attasami Diagnostic Center, Tripoli, Libya
| | - Rami Alazab
- Division of Urology, Jordan University of Science and Technology, Irbid, Irbid
| | - Kiril Trpkov
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
14
|
Kamdar S, Isserlin R, Van der Kwast T, Zlotta AR, Bader GD, Fleshner NE, Bapat B. Exploring targets of TET2-mediated methylation reprogramming as potential discriminators of prostate cancer progression. Clin Epigenetics 2019; 11:54. [PMID: 30917865 PMCID: PMC6438015 DOI: 10.1186/s13148-019-0651-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/10/2019] [Indexed: 12/13/2022] Open
Abstract
Background Global DNA methylation alterations are hallmarks of cancer. The tumor-suppressive TET enzymes, which are involved in DNA demethylation, are decreased in prostate cancer (PCa); in particular, TET2 is specifically targeted by androgen-dependent mechanisms of repression in PCa and may play a central role in carcinogenesis. Thus, the identification of key genes targeted by TET2 dysregulation may provide further insight into cancer biology. Results Using a CRISPR/Cas9-derived TET2-knockout prostate cell line, and through whole-transcriptome and whole-methylome sequencing, we identified seven candidate genes—ASB2, ETNK2, MEIS2, NRG1, NTN1, NUDT10, and SRPX—exhibiting reduced expression and increased promoter methylation, a pattern characteristic of tumor suppressors. Decreased expression of these genes significantly discriminates between recurrent and non-recurrent prostate tumors from the Cancer Genome Atlas (TCGA) cohort (n = 423), and ASB2, NUDT10, and SRPX were significantly correlated with lower recurrence-free survival in patients by Kaplan-Meier analysis. ASB2, MEIS2, and SRPX also showed significantly lower expression in high-risk Gleason score 8 tumors as compared to low or intermediate risk tumors, suggesting that these genes may be particularly useful as indicators of PCa progression. Furthermore, methylation array probes in the TCGA dataset, which were proximal to the highly conserved, differentially methylated sites identified in our TET2-knockout cells, were able to significantly distinguish between matched prostate tumor and normal prostate tissues (n = 50 pairs). Except ASB2, all genes exhibited significantly increased methylation at these probes, and methylation status of at least one probe for each of these genes showed association with measures of PCa progression such as recurrence, stage, or Gleason score. Since ASB2 did not have any probes within the TET2-knockout differentially methylated region, we validated ASB2 methylation in an independent series of matched tumor-normal samples (n = 19) by methylation-specific qPCR, which revealed concordant and significant increases in promoter methylation within the TET2-knockout site. Conclusions Our study identifies seven genes governed by TET2 loss in PCa which exhibit an association between their methylation and expression status and measures of PCa progression. As differential methylation profiles and TET2 expression are associated with advanced PCa, further investigation of these specialized TET2 targets may provide important insights into patterns of carcinogenic gene dysregulation. Electronic supplementary material The online version of this article (10.1186/s13148-019-0651-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shivani Kamdar
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 60 Murray Street, L6-304B, Toronto, ON, M5T 3L9, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building (6th floor), 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Ruth Isserlin
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College St, Toronto, ON, M5S 3E1, Canada
| | - Theodorus Van der Kwast
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building (6th floor), 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.,Department of Pathology, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, ON, M5G 2C4, Canada
| | - Alexandre R Zlotta
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 60 Murray Street, L6-304B, Toronto, ON, M5T 3L9, Canada.,Department of Surgery and Surgical Oncology, Division of Urology, University Health Network, University of Toronto, 190 Elizabeth St, Toronto, ON, M5G 2C4, Canada
| | - Gary D Bader
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College St, Toronto, ON, M5S 3E1, Canada
| | - Neil E Fleshner
- Department of Surgery and Surgical Oncology, Division of Urology, University Health Network, University of Toronto, 190 Elizabeth St, Toronto, ON, M5G 2C4, Canada
| | - Bharati Bapat
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 60 Murray Street, L6-304B, Toronto, ON, M5T 3L9, Canada. .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building (6th floor), 1 King's College Circle, Toronto, ON, M5S 1A8, Canada. .,Department of Pathology, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, ON, M5G 2C4, Canada. .,Department of Surgery and Surgical Oncology, Division of Urology, University Health Network, University of Toronto, 190 Elizabeth St, Toronto, ON, M5G 2C4, Canada.
| |
Collapse
|
15
|
Magi-Galluzzi C, Isharwal S, Falzarano SM, Tsiatis A, Dee A, Maddala T, Knezevic D, Febbo PG, Lawrence J, Klein EA. The 17-Gene Genomic Prostate Score Assay Predicts Outcome After Radical Prostatectomy Independent of PTEN Status. Urology 2018; 121:132-138. [PMID: 30142405 DOI: 10.1016/j.urology.2018.07.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 06/17/2018] [Accepted: 07/16/2018] [Indexed: 10/28/2022]
Abstract
OBJECTIVE To compare the ability of loss of phosphatase and tensin homolog (PTEN) and Genomic prostate score assay (GPS) in predicting the biochemical-recurrence (BCR) and clinical-recurrence (CR) after radical prostatectomy (RP) for clinically localized prostate cancer (PCa). METHODS Three hundred seventy seven patients with and without CR were retrospectively selected by stratified cohort sampling design from RP database. PTEN status (by immunohistochemistry [IHC] and fluorescence in situ hybridization [FISH]) and GPS results were determined for RP specimens. BCR was defined as Prostate Specific Antigen (PSA) ≥ 0.2 ng/mL or initiation of salvage therapy for a rising PSA. CR was defined as local recurrence and/or distant metastases. RESULTS Baseline mean age, PSA, and GPS score for the cohort were 61.1 years, 8 ng/dL, and 32.8. PTEN loss was noted in 38% patients by FISH and 25% by IHC. The concordance between FISH and IHC for PTEN loss was 66% (Kappa coefficient 0.278; P < .001). On univariable analysis, loss of PTEN by FISH or IHC was associated with BCR and CR (P < .05). However, after adjusting for GPS results, PTEN loss was not a significant predictor for CR or BCR (P > .1). The GPS result remained strongly associated with CR and BCR after adjusting for PTEN status (P < .001). PTEN status and GPS results only weakly correlated. GPS was widely distributed regardless of PTEN status indicating the biological heterogeneity of PCa even in PTEN-deficient cases. CONCLUSION GPS is a significant predictor of aggressive PCa, independent of PTEN status. After adjustment for GPS results, PTEN was not independently associated with recurrence for PCa.
Collapse
Affiliation(s)
- Cristina Magi-Galluzzi
- Department of Urology, Glickman Urology and Kidney Institute, Cleveland Clinic, Cleveland, OH; Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Sudhir Isharwal
- Department of Urology, Glickman Urology and Kidney Institute, Cleveland Clinic, Cleveland, OH
| | - Sara M Falzarano
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | | | - Anne Dee
- Genomic Health, Inc., Redwood City, CA
| | | | | | | | | | - Eric A Klein
- Department of Urology, Glickman Urology and Kidney Institute, Cleveland Clinic, Cleveland, OH.
| |
Collapse
|
16
|
Rao K, Liang S, Cardamone M, Joshu CE, Marmen K, Bhavsar N, Nelson WG, Ballentine Carter H, Albert MC, Platz EA, Pollack CE. Cost implications of PSA screening differ by age. BMC Urol 2018; 18:38. [PMID: 29743049 PMCID: PMC5944051 DOI: 10.1186/s12894-018-0344-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/19/2018] [Indexed: 11/21/2022] Open
Abstract
Background Multiple guidelines seek to alter rates of prostate-specific antigen (PSA)-based prostate cancer screening. The costs borne by payers associated with PSA-based screening for men of different age groups—including the costs of screening and subsequent diagnosis, treatment, and adverse events—remain uncertain. We sought to develop a model of PSA costs that could be used by payers and health care systems to inform cost considerations under a range of different scenarios. Methods We determined the prevalence of PSA screening among men aged 50 and higher using 2013-2014 data from a large, multispecialty group, obtained reimbursed costs associated with screening, diagnosis, and treatment from a commercial health plan, and identified transition probabilities for biopsy, diagnosis, treatment, and complications from the literature to generate a cost model. We estimated annual total costs for groups of men ages 50-54, 55-69, and 70+ years, and varied annual prostate cancer screening prevalence in each group from 5 to 50% and tested hypothetical examples of different test characteristics (e.g., true/false positive rate). Results Under the baseline screening patterns, costs of the PSA screening represented 10.1% of the total costs; costs of biopsies and associated complications were 23.3% of total costs; and, although only 0.3% of all screen eligible patients were treated, they accounted for 66.7% of total costs. For each 5-percentage point decrease in PSA screening among men aged 70 and older for a single calendar year, total costs associated with prostate cancer screening decreased by 13.8%. For each 5-percentage point decrease in PSA screening among men 50-54 and 55-69 years old, costs were 2.3% and 7.3% lower respectively. Conclusions With constrained financial resources and with national pressure to decrease use of clinically unnecessary PSA-based prostate cancer screening, there is an opportunity for cost savings, especially by focusing on the downstream costs disproportionately associated with screening men 70 and older. Electronic supplementary material The online version of this article (10.1186/s12894-018-0344-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Karthik Rao
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stella Liang
- Financial Analysis Unit, Johns Hopkins Health System, Baltimore, MD, USA
| | - Michael Cardamone
- Financial Analysis Unit, Johns Hopkins Health System, Baltimore, MD, USA
| | - Corinne E Joshu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Kyle Marmen
- Johns Hopkins Health Care, Glen Burnie, Baltimore, MD, USA
| | - Nrupen Bhavsar
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - William G Nelson
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - H Ballentine Carter
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael C Albert
- Johns Hopkins Community Physicians, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Elizabeth A Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Craig E Pollack
- Department of Medicine, Johns Hopkins University School of Medicine, 2024 E. Monument Street, Suite 2-519, Baltimore, MD, 21287, USA.
| |
Collapse
|
17
|
Prostate Cancer Genomics: Recent Advances and the Prevailing Underrepresentation from Racial and Ethnic Minorities. Int J Mol Sci 2018; 19:ijms19041255. [PMID: 29690565 PMCID: PMC5979433 DOI: 10.3390/ijms19041255] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 04/15/2018] [Accepted: 04/15/2018] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (CaP) is the most commonly diagnosed non-cutaneous cancer and the second leading cause of male cancer deaths in the United States. Among African American (AA) men, CaP is the most prevalent malignancy, with disproportionately higher incidence and mortality rates. Even after discounting the influence of socioeconomic factors, the effect of molecular and genetic factors on racial disparity of CaP is evident. Earlier studies on the molecular basis for CaP disparity have focused on the influence of heritable mutations and single-nucleotide polymorphisms (SNPs). Most CaP susceptibility alleles identified based on genome-wide association studies (GWAS) were common, low-penetrance variants. Germline CaP-associated mutations that are highly penetrant, such as those found in HOXB13 and BRCA2, are usually rare. More recently, genomic studies enabled by Next-Gen Sequencing (NGS) technologies have focused on the identification of somatic mutations that contribute to CaP tumorigenesis. These studies confirmed the high prevalence of ERG gene fusions and PTEN deletions among Caucasian Americans and identified novel somatic alterations in SPOP and FOXA1 genes in early stages of CaP. Individuals with African ancestry and other minorities are often underrepresented in these large-scale genomic studies, which are performed primarily using tumors from men of European ancestry. The insufficient number of specimens from AA men and other minority populations, together with the heterogeneity in the molecular etiology of CaP across populations, challenge the generalizability of findings from these projects. Efforts to close this gap by sequencing larger numbers of tumor specimens from more diverse populations, although still at an early stage, have discovered distinct genomic alterations. These research findings can have a direct impact on the diagnosis of CaP, the stratification of patients for treatment, and can help to address the disparity in incidence and mortality of CaP. This review examines the progress of understanding in CaP genetics and genomics and highlight the need to increase the representation from minority populations.
Collapse
|
18
|
Yajun C, Yuan T, Zhong W, Bin X. Investigation of the molecular mechanisms underlying postoperative recurrence in prostate cancer by gene expression profiling. Exp Ther Med 2018; 15:761-768. [PMID: 29399083 PMCID: PMC5772610 DOI: 10.3892/etm.2017.5510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 10/20/2017] [Indexed: 12/30/2022] Open
Abstract
The present study aimed to identify potential genes associated with prostate cancer (PCa) recurrence following radical prostatectomy (RP) in order to improve the prediction of the prognosis of patients with PCa. The GSE25136 microarray dataset, including 39 recurrent and 40 non-recurrent PCa samples, was downloaded from the Gene Expression Omnibus database. Differentially-expressed genes (DEGs) were identified using limma packages, and the pheatmap package was used to present the DEGs screened using a hierarchical cluster analysis. Furthermore, gene ontology functional enrichment analysis was used to predict the potential functions of the DEGs. Subsequently, Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were performed to analyze pathway enrichment of DEGs in the regulatory network. Lastly, a protein-protein interaction (PPI) network of the DEGs was constructed using Cytoscape software to understand the interactions between these DEGs. A total of 708 DEGs were identified in the recurrent and non-recurrent PCa samples. Functional annotation revealed that these DEGs were primarily involved in cell adhesion, negative regulation of growth, and the cyclic adenosine monophosphate and mitogen-activated protein kinase (MAPK) signaling pathways. Furthermore, five key genes, including cluster of differentiation 22, insulin-like growth factor-1, inhibin β A subunit, MAPK kinase 5 and receptor tyrosine kinase like orphan receptor 1, were identified through PPI network analysis. The results of the present study have provided novel ideas for predicting the prognosis of patients with PCa following RP.
Collapse
Affiliation(s)
- Cheng Yajun
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Tang Yuan
- Department of Gastrointestinal Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Wang Zhong
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Xu Bin
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
19
|
Fujimura T, Takayama K, Takahashi S, Inoue S. Estrogen and Androgen Blockade for Advanced Prostate Cancer in the Era of Precision Medicine. Cancers (Basel) 2018; 10:cancers10020029. [PMID: 29360794 PMCID: PMC5836061 DOI: 10.3390/cancers10020029] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/19/2018] [Accepted: 01/19/2018] [Indexed: 12/14/2022] Open
Abstract
Androgen deprivation therapy (ADT) has been widely prescribed for patients with advanced prostate cancer (PC) to control key signaling pathways via androgen receptor (AR) and AR-collaborative transcriptional factors; however, PC gradually acquires a lethal phenotype and results in castration-resistant PC (CRPC) during ADT. Therefore, new therapeutic strategies are required in clinical practice. In addition, ARs; estrogen receptors (ERs; ERα and ERβ); and estrogen-related receptors (ERRs; ERRα, ERRβ, and ERRγ) have been reported to be involved in the development or regulation of PC. Recent investigations have revealed the role of associated molecules, such as KLF5, FOXO1, PDGFA, VEGF-A, WNT5A, TGFβ1, and micro-RNA 135a of PC, via ERs and ERRs. Selective ER modulators (SERMs) have been developed. Recently, estrogen and androgen blockade (EAB) using a combination of toremifene and ADT has been demonstrated to improve biochemical recurrence rate in treatment-naïve bone metastatic PC. In the future, the suitability of ADT alone or EAB for individuals may be evaluated by making clinical decisions on the basis of information obtained from RT-PCR, gene-panel, or liquid biopsy to create a “personalized medicine” or “precision medicine”. In this review, we summarize ER and ERR signaling pathways, molecular diagnosis, and SERMs as candidates for advanced PC treatment.
Collapse
Affiliation(s)
- Tetsuya Fujimura
- Department of Urology, National Center for Global Health and Medicine, Tokyo 162-8655, Japan.
| | - Kenichi Takayama
- Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan.
| | - Satoru Takahashi
- Department of Urology, Nihon University School of Medicine, Tokyo 173-8610, Japan.
| | - Satoshi Inoue
- Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan.
| |
Collapse
|
20
|
Li H, Wang Z, Xiao W, Yan L, Guan W, Hu Z, Wu L, Huang Q, Wang J, Xu H, Zhang X, Ye Z. Androgen-receptor splice variant-7-positive prostate cancer: a novel molecular subtype with markedly worse androgen-deprivation therapy outcomes in newly diagnosed patients. Mod Pathol 2018; 31:198-208. [PMID: 29076496 DOI: 10.1038/modpathol.2017.74] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 05/15/2017] [Indexed: 12/31/2022]
Abstract
Androgen-deprivation therapy has been the standard treatment for metastatic and locally advanced prostate cancer, but the majority of patients will progress to castration-resistant prostate cancer within 2-3 years. Unlike the case in breast cancer, no clinically validated biomarker has been used to predict the outcomes of androgen-deprivation therapy. To evaluate androgen-receptor splice variant-7 (AR-V7) detection in newly diagnosed advanced prostate cancer and describe the distinctive prognosis of this novel molecular subtype, this study retrospectively enrolled 168 newly diagnosed prostate cancer patients from 2003 to 2015 who received androgen-deprivation therapy. AR-V7 immunohistochemical staining was performed with a monoclonal antibody, and AR-V7 expression was determined using Immune-Reactive Score data. The association between nuclear AR-V7 expression and prognosis was determined. Multiple cause-specific Cox regression and stratified cumulative incidences were used to analyze the prognosis risk. Among the 168 patients, 32 (19%) were AR-V7-positive. Compared with the AR-V7-negative patients, the AR-V7-positive patients had significantly lower prostate-specific antigen response rates (P<0.001) to androgen-deprivation therapy and a much shorter time to castration-resistant prostate cancer (P<0.0001). In Kaplan-Meier analysis, the AR-V7-positive group showed markedly lower castration-resistant prostate cancer progression-free survival (P<0.0001) and much lower cancer-specific (P<0.0001) and overall survival (P<0.0001) both in all enrolled patients and in patients with metastases. AR-V7 positivity was a significant predictor of castration-resistant prostate cancer progression in multiple Cox regression (hazard ratio: 4.826; 95% CI: 2.960-7.869; P<0.001). AR-V7 immunohistochemical detection in newly diagnosed prostate cancer patients who are planning to receive androgen-deprivation therapy, especially those with metastases, is necessary and valuable for prognostic assessment. AR-V7-positive prostate cancer should be considered a novel prostate cancer subtype that should be distinguished upon initial biopsy. The main limitation of this study is its observational nature.
Collapse
Affiliation(s)
- Heng Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhize Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Xiao
- Translational Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Libin Yan
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Guan
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiquan Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lily Wu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | - Ji Wang
- Department of Cell Death and Cancer Genetics, the Hormel Institute University of Minnesota, Austin, USA
| | - Hua Xu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xu Zhang
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital/PLA Medical School, Beijing, China
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Qi X, Wang Y, Hou J, Huang Y. A Single Nucleotide Polymorphism in HPGD Gene Is Associated with Prostate Cancer Risk. J Cancer 2017; 8:4083-4086. [PMID: 29187884 PMCID: PMC5706011 DOI: 10.7150/jca.22025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 08/26/2017] [Indexed: 01/18/2023] Open
Abstract
Introduction: The HPGD gene was associated with some cancers, such as colorectal, breast, prostate, and bladder. However, detailed role of 15-hydroxyprostaglandin dehydrogenase (HPGD) gene remain unclear in prostate cancer. The study was to investigate the correlation between rs8752 that located in the 3'untranslated region (UTR) of the 15-hydroxyprostaglandin dehydrogenase (HPGD) gene and prostate cancer (PCa) risk. Materials and Methods: 109 patients from the First Affiliate Hospital of Soochow University were recruited. According to the results of pathologic diagnosis, all patients were divided into two groups (prostate cancer and benign prostatic hyperplasia). The single-nucleotide polymorphism (SNP) rs8752 was genotyped in all samples by direct sequencing. Results: 54 prostate cancer and 55 BPH patients were included with a median age of 70.41 and 67.62 years, respectively. No statistically significant difference between two groups in patient criteria. The frequency of the GG homozygote and AG+GG genotype were 37.74% and 62.26% in 54 prostate cancer samples, while in 55BPH patients, values were 62.50% and 37.50%. Compared with the GG genotype, the combined GA+AA genotypes had a significantly higher risk of prostate cancer (OR = 2.750; 95% CI: 1.266-5.971, p = 0.011). Furthermore, the risk effect was obtained in subgroups of PCa patient group, the AA+AG genotypes significantly associated with the higher Gleason score samples (AA+AG vs GG: OR = 3.50, 95%CI = 1.106-11.072, p = 0.033) and the risk of pathological stage (AA+AG vs GG: OR = 4.00, 95%CI = 1.253-12.767, p = 0.019). Conclusions: rs8752 in the 3'untranslated region (UTR) of the 15-hydroxyprostaglandin dehydrogenase (HPGD) gene was found to be responsible for the susceptibility to prostate cancer in Chinese individuals.
Collapse
Affiliation(s)
- Xiaofei Qi
- Department of Urology, the First Affiliate Hospital of Soochow University, Suzhou, China
| | - Yu Wang
- Department of Urology, the First Affiliate Hospital of Soochow University, Suzhou, China
| | - Jianquan Hou
- Department of Urology, the First Affiliate Hospital of Soochow University, Suzhou, China
| | - Yuhua Huang
- Department of Urology, the First Affiliate Hospital of Soochow University, Suzhou, China
| |
Collapse
|
22
|
Worst TS, Waldbillig F, Abdelhadi A, Weis CA, Gottschalt M, Steidler A, von Hardenberg J, Michel MS, Erben P. The EEF1A2 gene expression as risk predictor in localized prostate cancer. BMC Urol 2017; 17:86. [PMID: 28923030 PMCID: PMC5604352 DOI: 10.1186/s12894-017-0278-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 09/13/2017] [Indexed: 12/31/2022] Open
Abstract
Background Besides clinical stage and Gleason score, risk-stratification of prostate cancer in the pretherapeutic setting mainly relies on the serum PSA level. Yet, this is associated with many uncertainties. With regard to therapy decision-making, additional markers are needed to allow an exact risk prediction. Eukaryotic translation elongation factor 1 alpha 2 (EEF1A2) was previously suggested as driver of tumor progression and potential biomarker. In the present study its functional and prognostic relevance in prostate cancer was investigated. Methods EEF1A2 expression was analyzed in two cohorts of patients (n = 40 and n = 59) with localized PCa. Additionally data from two large expression dataset (MSKCC, Cell, 2010 with n = 131 localized, n = 19 metastatic PCa and TCGA provisional data, n = 499) of PCa patients were reanalyzed. The expression of EEF1A2 was correlated with histopathology features and biochemical recurrence (BCR). To evaluate the influence of EEF1A2 on proliferation and migration of metastatic PC3 cells, siRNA interference was used. Statistical significance was tested with t-test, Mann-Whitney-test, Pearson correlation and log-rank test. Results qRT-PCR revealed EEF1A2 to be significantly overexpressed in PCa tissue, with an increase according to tumor stage in one cohort (p = 0.0443). In silico analyses in the MSKCC cohort confirmed the overexpression of EEF1A2 in localized PCa with high Gleason score (p = 0.0142) and in metastatic lesions (p = 0.0038). Patients with EEF1A2 overexpression had a significantly shorter BCR-free survival (p = 0.0028). EEF1A2 expression was not correlated with serum PSA levels. Similar results were seen in the TCGA cohort, where EEF1A2 overexpression only occurred in tumors with Gleason 7 or higher. Patients with elevated EEF1A2 expression had a significantly shorter BCR-free survival (p = 0.043). EEF1A2 knockdown significantly impaired the migration, but not the proliferation of metastatic PC3 cells. Conclusion The overexpression of EEF1A2 is a frequent event in localized PCa and is associated with histopathology features and a shorter biochemical recurrence-free survival. Due to its independence from serum PSA levels, EEF1A2 could serve as valuable biomarker in risk-stratification of localized PCa.
Collapse
Affiliation(s)
- Thomas Stefan Worst
- Department of Urology, University Medical Centre Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany. .,Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Frank Waldbillig
- Department of Urology, University Medical Centre Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Abdallah Abdelhadi
- Department of Urology, University Medical Centre Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Cleo-Aron Weis
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Maria Gottschalt
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Annette Steidler
- Department of Urology, University Medical Centre Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Jost von Hardenberg
- Department of Urology, University Medical Centre Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Maurice Stephan Michel
- Department of Urology, University Medical Centre Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Philipp Erben
- Department of Urology, University Medical Centre Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to examine prostate cancer racial disparities specific to the African-American population. RECENT FINDINGS African-American men are more likely to be diagnosed with prostate cancer, present at an earlier age; are more likely to have locally advanced or metastatic disease at diagnosis; and have suboptimal outcomes to standard treatments. Prostate cancer treatment requires a nuanced approach, particularly when applying screening, counseling, and management of African-American men. Oncological as well as functional outcomes may differ and are potentially due to a combination of genetic, molecular, behavioral, and socioeconomic factors.
Collapse
Affiliation(s)
- Zachary L Smith
- Department of Surgery, Section of Urology, The University of Chicago Medicine, 5841 S. Maryland Avenue, MC 6038, Chicago, IL, 60637, USA.
| | - Scott E Eggener
- Department of Surgery, Section of Urology, The University of Chicago Medicine, 5841 S. Maryland Avenue, MC 6038, Chicago, IL, 60637, USA
| | - Adam B Murphy
- Department of Urology, Northwestern University Feinberg School of Medicine, Tarry Building Room 16-703, 300 E. Superior Street, Chicago, IL, 60611, USA
| |
Collapse
|
24
|
Wang Y, Freedman JA, Liu H, Moorman PG, Hyslop T, George DJ, Lee NH, Patierno SR, Wei Q. Associations between RNA splicing regulatory variants of stemness-related genes and racial disparities in susceptibility to prostate cancer. Int J Cancer 2017; 141:731-743. [PMID: 28510291 DOI: 10.1002/ijc.30787] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 04/24/2017] [Accepted: 05/02/2017] [Indexed: 01/01/2023]
Abstract
Evidence suggests that cells with a stemness phenotype play a pivotal role in oncogenesis, and prostate cells exhibiting this phenotype have been identified. We used two genome-wide association study (GWAS) datasets of African descendants, from the Multiethnic/Minority Cohort Study of Diet and Cancer (MEC) and the Ghana Prostate Study, and two GWAS datasets of non-Hispanic whites, from the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial and the Breast and Prostate Cancer Cohort Consortium (BPC3), to analyze the associations between genetic variants of stemness-related genes and racial disparities in susceptibility to prostate cancer. We evaluated associations of single-nucleotide polymorphisms (SNPs) in 25 stemness-related genes with prostate cancer risk in 1,609 cases and 2,550 controls of non-Hispanic whites (4,934 SNPs) and 1,144 cases and 1,116 controls of African descendants (5,448 SNPs) with correction by false discovery rate ≤0.2. We identified 32 SNPs in five genes (TP63, ALDH1A1, WNT1, MET and EGFR) that were significantly associated with prostate cancer risk, of which six SNPs in three genes (TP63, ALDH1A1 and WNT1) and eight EGFR SNPs showed heterogeneity in susceptibility between these two racial groups. In addition, 13 SNPs in MET and one in ALDH1A1 were found only in African descendants. The in silico bioinformatics analyses revealed that EGFR rs2072454 and SNPs in linkage with the identified SNPs in MET and ALDH1A1 (r2 > 0.6) were predicted to regulate RNA splicing. These variants may serve as novel biomarkers for racial disparities in prostate cancer risk.
Collapse
Affiliation(s)
- Yanru Wang
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC
| | - Jennifer A Freedman
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC
| | - Patricia G Moorman
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Community and Family Medicine, Duke University Medical Center, Durham, NC
| | - Terry Hyslop
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC
| | - Daniel J George
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC
| | - Norman H Lee
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Steven R Patierno
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC.,Department of Community and Family Medicine, Duke University Medical Center, Durham, NC.,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC
| |
Collapse
|
25
|
Chen Z, Gerke T, Bird V, Prosperi M. Trends in Gene Expression Profiling for Prostate Cancer Risk Assessment: A Systematic Review. Biomed Hub 2017; 2:1-15. [PMID: 31988908 PMCID: PMC6945900 DOI: 10.1159/000472146] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 03/07/2017] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES The aim of the study is to review biotechnology advances in gene expression profiling on prostate cancer (PCa), focusing on experimental platform development and gene discovery, in relation to different study designs and outcomes in order to understand how they can be exploited to improve PCa diagnosis and clinical management. METHODS We conducted a systematic literature review on gene expression profiling studies through PubMed/MEDLINE and Web of Science between 2000 and 2016. Tissue biopsy and clinical gene profiling studies with different outcomes (e.g., recurrence, survival) were included. RESULTS Over 3,000 papers were screened and 137 full-text articles were selected. In terms of technology used, microarray is still the most popular technique, increasing from 50 to 70% between 2010 and 2015, but there has been a rise in the number of studies using RNA sequencing (13% in 2015). Sample sizes have increased, as well as the number of genes that can be screened all at once, but we have also observed more focused targeting in more recent studies. Qualitative analysis on the specific genes found associated with PCa risk or clinical outcomes revealed a large variety of gene candidates, with a few consistent cross-studies. CONCLUSIONS The last 15 years of research in gene expression in PCa have brought a large volume of data and information that has been decoded only in part, but advancements in high-throughput sequencing technology are increasing the amount of data that can be generated. The variety of findings warrants the execution of both validation studies and meta-analyses. Genetic biomarkers have tremendous potential for early diagnosis of PCa and, if coupled with other diagnostics (e.g., imaging), can effectively be used to concretize less-invasive, personalized prediction of PCa risk and progression.
Collapse
Affiliation(s)
- Zhaoyi Chen
- Department of Epidemiology, College of Public Health and Health Professions, College of Medicine, University of Florida, Gainesville, FL, USA
| | | | - Victoria Bird
- Department of Urology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Mattia Prosperi
- Department of Epidemiology, College of Public Health and Health Professions, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
26
|
Worst TS, von Hardenberg J, Gross JC, Erben P, Schnölzer M, Hausser I, Bugert P, Michel MS, Boutros M. Database-augmented Mass Spectrometry Analysis of Exosomes Identifies Claudin 3 as a Putative Prostate Cancer Biomarker. Mol Cell Proteomics 2017; 16:998-1008. [PMID: 28396511 DOI: 10.1074/mcp.m117.068577] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/23/2017] [Indexed: 01/01/2023] Open
Abstract
In prostate cancer and other malignancies sensitive and robust biomarkers are lacking or have relevant limitations. Prostate specific antigen (PSA), the only biomarker widely used in prostate cancer, is suffering from low specificity. Exosomes offer new perspectives in the discovery of blood-based biomarkers. Here we present a proof-of principle study for a proteomics-based identification pipeline, implementing existing data sources, to exemplarily identify exosome-based biomarker candidates in prostate cancer.Exosomes from malignant PC3 and benign PNT1A cells and from FBS-containing medium were isolated using sequential ultracentrifugation. Exosome and control samples were analyzed on an LTQ-Orbitrap XL mass spectrometer. Proteomic data is available via ProteomeXchange with identifier PXD003651. We developed a scoring scheme to rank 64 proteins exclusively found in PC3 exosomes, integrating data from four public databases and published mass spectrometry data sets. Among the top candidates, we focused on the tight junction protein claudin 3. Retests under serum-free conditions using immunoblotting and immunogold labeling confirmed the presence of claudin 3 on PC3 exosomes. Claudin 3 levels were determined in the blood plasma of patients with localized (n = 58; 42 with Gleason score 6-7, 16 with Gleason score ≥8) and metastatic prostate cancer (n = 11) compared with patients with benign prostatic hyperplasia (n = 15) and healthy individuals (n = 15) using ELISA, without prior laborious exosome isolation. ANOVA showed different CLDN3 plasma levels in these groups (p = 0.004). CLDN3 levels were higher in patients with Gleason ≥8 tumors compared with patients with benign prostatic hyperplasia (p = 0.012) and Gleason 6-7 tumors (p = 0.029). In patients with localized tumors CLDN3 levels predicted a Gleason score ≥ 8 (AUC = 0.705; p = 0.016) and did not correlate with serum PSA.By using the described workflow claudin 3 was identified and validated as a potential blood-based biomarker in prostate cancer. Furthermore this workflow could serve as a template to be used in other cancer entities.
Collapse
Affiliation(s)
- Thomas Stefan Worst
- From the ‡Department of Urology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; .,§Department of Signaling and Functional Genomics, German Cancer Research Center, Heidelberg, Germany
| | - Jost von Hardenberg
- From the ‡Department of Urology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,§Department of Signaling and Functional Genomics, German Cancer Research Center, Heidelberg, Germany
| | - Julia Christina Gross
- §Department of Signaling and Functional Genomics, German Cancer Research Center, Heidelberg, Germany.,¶Haematology and Oncology and Developmental Biochemistry, University Medical Center, Göttingen, Germany
| | - Philipp Erben
- From the ‡Department of Urology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Martina Schnölzer
- ‖Genomics and Proteomics Core Facility, German Cancer Research Center, Heidelberg, Germany
| | - Ingrid Hausser
- **Institute of Pathology IPH, University Clinic Heidelberg and Electron Microscopy Core Facility, University of Heidelberg, Germany
| | - Peter Bugert
- ‡‡Institute of Transfusion Medicine and Immunology, Heidelberg University, Medical Faculty Mannheim, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Maurice Stephan Michel
- From the ‡Department of Urology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Michael Boutros
- §Department of Signaling and Functional Genomics, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
27
|
Hu AX, Huang ZY, Zhang L, Shen J. Potential prognostic long non-coding RNA identification and their validation in predicting survival of patients with multiple myeloma. Tumour Biol 2017; 39:1010428317694563. [PMID: 28378636 DOI: 10.1177/1010428317694563] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Multiple myeloma, a typical hematological malignancy, is characterized by malignant proliferation of plasma cells. This study was to identify differently expressed long non-coding RNAs to predict the survival of patients with multiple myeloma efficiently. Gene expressing profiles of diagnosed patients with multiple myeloma, GSE24080 (559 samples) and GSE57317 (55 samples), were downloaded from Gene Expression Omnibus database. After processing, survival-related long non-coding RNAs were identified by Cox regression analysis. The prognosis of multiple myeloma patients with differently expressed long non-coding RNAs was predicted by Kaplan–Meier analysis. Meanwhile, stratified analysis was performed based on the concentrations of serum beta 2-microglobulin (S-beta 2m), albumin, and lactate dehydrogenase of multiple myeloma patients. Gene set enrichment analysis was performed to further explore the functions of identified long non-coding RNAs. A total of 176 long non-coding RNAs significantly related to the survival of multiple myeloma patients (p < 0.05) were identified. In dataset GSE24080 and GSE57317, there were 558 and 55 patients being clustered into two groups with significant differences, respectively. Stratified analysis indicated that prediction of the prognoses with these long non-coding RNAs was independent from other clinical phenotype of multiple myeloma. Gene set enrichment analysis–identified pathways of cell cycle, focal adhesion, and G2-M checkpoint were associated with these long non-coding RNAs. A total of 176 long non-coding RNAs, especially RP1-286D6.1, AC008875.2, MTMR9L, AC069360.2, and AL512791.1, were potential biomarkers to evaluate the prognosis of multiple myeloma patients. These long non-coding RNAs participated indispensably in many pathways associated to the development of multiple myeloma; however, the molecular mechanisms need to be further studied.
Collapse
Affiliation(s)
- Ai-Xin Hu
- Department of Orthopedic Surgery, People’s Hospital of Three Gorges University, Yichang, China
| | - Zhi-Yong Huang
- PuAi Institute, Edong Healthcare Group, Huangshi Central Hospital, Huangshi, China
| | - Lin Zhang
- Department of Spinal Surgery, The Affiliated Huai’an Hospital of Xuzhou Medical University and The Second People’s Hospital of Huai’an, Huai’an, China
| | - Jian Shen
- Changzhou Hygiene Vocational Technology School, Changzhou, China
| |
Collapse
|
28
|
Analysis of Argonaute Complex Bound mRNAs in DU145 Prostate Carcinoma Cells Reveals New miRNA Target Genes. Prostate Cancer 2017; 2017:4893921. [PMID: 28163933 PMCID: PMC5253174 DOI: 10.1155/2017/4893921] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 10/30/2016] [Indexed: 12/14/2022] Open
Abstract
Posttranscriptional gene regulation by microRNAs (miRNAs) contributes to the induction and maintenance of prostate carcinoma (PCa). To identify mRNAs enriched or removed from Ago2-containing RISC complexes, these complexes were immunoprecipitated from normal prostate fibroblasts (PNFs) and the PCa line DU145 and the bound mRNAs were quantified by microarray. The analysis of Ago complexes derived from PNFs or DU145 confirmed the enrichment or depletion of a variety of mRNAs already known from the literature to be deregulated. Novel potential targets were analyzed by luciferase assays with miRNAs known to be deregulated in PCa. We demonstrate that the mRNAs of the death effector domain-containing protein (DEDD), the tumor necrosis factor receptor superfamily, member 10b protein (TNFRSF10B), the tumor protein p53 inducible nuclear protein 1 (TP53INP1), and the secreted protein, acidic, cysteine-rich (SPARC; osteonectin) are regulated by miRNAs miR-148a, miR-20a, miR-24, and miR-29a/b, respectively. Therefore, these miRNAs represent potential targets for therapy. Surprisingly, overexpression of miR-24 induced focus formation and proliferation of DU145 cells, while miR-29b reduced proliferation. The study confirms genes deregulated in PCa by virtue of their presence/absence in the Ago2-complex. In conjunction with the already published miRNA profiles of PCa, the data can be used to identify miRNA-regulated mRNAs.
Collapse
|
29
|
Albright FS, Stephenson RA, Agarwal N, Cannon-Albright LA. Relative Risks for Lethal Prostate Cancer Based on Complete Family History of Prostate Cancer Death. Prostate 2017; 77:41-48. [PMID: 27527734 DOI: 10.1002/pros.23247] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 08/02/2016] [Indexed: 12/27/2022]
Abstract
BACKGROUND There are few published familial relative risks (RR) for lethal prostate cancer. This study estimates RRs for lethal prostate cancer based on comprehensive family history data, with the goal of improving identification of those men at highest risk of dying from prostate cancer. METHODS We used a population-based genealogical resource linked to a statewide electronic SEER cancer registry and death certificates to estimate relative risks (RR) for death from prostate cancer based upon family history. Over 600,000 male probands were analyzed, representing a variety of family history constellations of lethal prostate cancer. RR estimates were based on the ratio of the observed to the expected number of lethal prostate cancer cases using internal rates. RESULTS RRs for lethal prostate cancer based on the number of affected first-degree relatives (FDR) ranged from 2.49 (95% CI: 2.27, 2.73) for exactly 1 FDR to 5.30 (2.13, 10.93) for ≥3 affected FDRs. In an absence of affected FDRs, increased risk was also significant for increasing numbers of affected second-degree or third degree relatives. Equivalent risks were observed for similar maternal and paternal family history. CONCLUSIONS This study provides population-based estimates of lethal prostate cancer risk based on lethal prostate cancer family history. Many family history constellations associated with two to greater than five times increased risk for lethal prostate cancer were identified. These lethal prostate cancer risk estimates hold potential for use in identification, screening, early diagnosis, and treatment of men at high risk for death from prostate cancer. Prostate77:41-48, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Frederick S Albright
- Pharmacotherapy Outcomes Research Center, Department of Pharmacotherapy, College of Pharmacy, University of Utah, Salt Lake City, Utah
| | - Robert A Stephenson
- Huntsman Cancer Institute, Salt Lake City, Utah
- Division of Urology, Department of Surgery, School of Medicine, University of Utah, Salt Lake City, Utah
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Neeraj Agarwal
- Huntsman Cancer Institute, Salt Lake City, Utah
- Division of Medical Oncology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| | - Lisa A Cannon-Albright
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah
- Division of Genetic Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
30
|
Zhao S, Geybels MS, Leonardson A, Rubicz R, Kolb S, Yan Q, Klotzle B, Bibikova M, Hurtado-Coll A, Troyer D, Lance R, Lin DW, Wright JL, Ostrander EA, Fan JB, Feng Z, Stanford JL. Epigenome-Wide Tumor DNA Methylation Profiling Identifies Novel Prognostic Biomarkers of Metastatic-Lethal Progression in Men Diagnosed with Clinically Localized Prostate Cancer. Clin Cancer Res 2017; 23:311-319. [PMID: 27358489 PMCID: PMC5199634 DOI: 10.1158/1078-0432.ccr-16-0549] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 06/08/2016] [Accepted: 06/11/2016] [Indexed: 01/22/2023]
Abstract
PURPOSE Aside from Gleason sum, few factors accurately identify the subset of prostate cancer patients at high risk for metastatic progression. We hypothesized that epigenetic alterations could distinguish prostate tumors with life-threatening potential. EXPERIMENTAL DESIGN Epigenome-wide DNA methylation profiling was performed in surgically resected primary tumor tissues from a population-based (n = 430) and a replication (n = 80) cohort of prostate cancer patients followed prospectively for at least 5 years. Metastasis was confirmed by positive bone scan, MRI, CT, or biopsy, and death certificates confirmed cause of death. AUC, partial AUC (pAUC, 95% specificity), and P value criteria were used to select differentially methylated CpG sites that robustly stratify patients with metastatic-lethal from nonrecurrent tumors, and which were complementary to Gleason sum. RESULTS Forty-two CpG biomarkers stratified patients with metastatic-lethal versus nonrecurrent prostate cancer in the discovery cohort, and eight of these CpGs replicated in the validation cohort based on a significant (P < 0.05) AUC (range, 0.66-0.75) or pAUC (range, 0.007-0.009). The biomarkers that improved discrimination of patients with metastatic-lethal prostate cancer include CpGs in five genes (ALKBH5, ATP11A, FHAD1, KLHL8, and PI15) and three intergenic regions. In the validation dataset, the AUC for Gleason sum alone (0.82) significantly increased with the addition of four individual CpGs (range, 0.86-0.89; all P <0.05). CONCLUSIONS Eight differentially methylated CpGs that distinguish patients with metastatic-lethal from nonrecurrent tumors were validated. These novel epigenetic biomarkers warrant further investigation as they may improve prognostic classification of patients with clinically localized prostate cancer and provide new insights on tumor aggressiveness. Clin Cancer Res; 23(1); 311-9. ©2016 AACR.
Collapse
Affiliation(s)
- Shanshan Zhao
- National Institute of Environmental Health Sciences, Biostatistics and Computational Biology Branch, Research Triangle Park, Durham, North Carolina
| | - Milan S Geybels
- Division of Public Health Sciences, Fred Hutchison Cancer Research Center, Seattle, Washington
- Department of Epidemiology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Amy Leonardson
- Division of Public Health Sciences, Fred Hutchison Cancer Research Center, Seattle, Washington
| | - Rohina Rubicz
- Division of Public Health Sciences, Fred Hutchison Cancer Research Center, Seattle, Washington
| | - Suzanne Kolb
- Division of Public Health Sciences, Fred Hutchison Cancer Research Center, Seattle, Washington
| | - Qingxiang Yan
- MD Anderson Cancer Center, Department of Biostatistics, Houston, Texas
| | | | | | - Antonio Hurtado-Coll
- Department of Urologic Sciences, University of British Columbia, and the Prostate Center, Vancouver General Hospital, Vancouver, Canada
| | - Dean Troyer
- Departments of Pathology, Microbiology, and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia
| | - Raymond Lance
- Department of Urology, Eastern Virginia Medical School, Norfolk, Virginia
| | - Daniel W Lin
- Division of Public Health Sciences, Fred Hutchison Cancer Research Center, Seattle, Washington
- Department of Urology, University of Washington School of Medicine, Seattle, Washington
| | - Jonathan L Wright
- Division of Public Health Sciences, Fred Hutchison Cancer Research Center, Seattle, Washington
- Department of Urology, University of Washington School of Medicine, Seattle, Washington
| | - Elaine A Ostrander
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland
| | | | - Ziding Feng
- MD Anderson Cancer Center, Department of Biostatistics, Houston, Texas
| | - Janet L Stanford
- Division of Public Health Sciences, Fred Hutchison Cancer Research Center, Seattle, Washington.
- Department of Epidemiology, University of Washington School of Public Health, Seattle, Washington
| |
Collapse
|
31
|
Benafif S, Eeles R. Genetic predisposition to prostate cancer. Br Med Bull 2016; 120:75-89. [PMID: 27941040 DOI: 10.1093/bmb/ldw039] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 08/31/2016] [Accepted: 09/04/2016] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Prostate cancer (PrCa) is the commonest non-cutaneous cancer in men in the UK. Epidemiological evidence as well as twin studies points towards a genetic component contributing to aetiology. SOURCES OF DATA Key recently published literature. AREAS OF AGREEMENT A family history of PrCa doubles the risk of disease development in first-degree relatives. Linkage and genetic sequencing studies identified rare moderate-high-risk gene loci, which predispose to PrCa development when altered by mutation. Genome-wide association studies have identified common single-nucleotide polypmorphisms (SNPs), which confer a cumulative risk of PrCa development with increasing number of risk alleles. There are emerging data that castrate-resistant disease is associated with mutations in DNA repair genes. AREAS OF CONTROVERSY Linkage studies investigating possible high-risk loci leading to PrCa development identified possible loci on several chromosomes, but most have not been consistently replicated by subsequent studies. Germline SNPs related to prostate specific antigen (PSA) levels and to normal tissue radiosensitivity have also been identified though not all have been validated in subsequent studies. GROWING POINTS Utilizing germline SNP profiles as well as identifying high-risk genetic variants could target screening to high-risk groups, avoiding the drawbacks of PSA screening. AREAS TIMELY FOR DEVELOPING RESEARCH Incorporating genetics into PrCa screening is being investigated currently using both common SNP profiles and higher risk rare variants. Knowledge of germline genetic defects will allow the development of targeted screening programs, preventive strategies and the personalized treatment of PrCa.
Collapse
Affiliation(s)
- S Benafif
- Institute of Cancer Research, Oncogenetics, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, London, UK
| | - R Eeles
- Institute of Cancer Research, Oncogenetics, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, London, UK
| |
Collapse
|
32
|
Molecular Markers and Targeted Therapeutics in Metastatic Tumors of the Spine: Changing the Treatment Paradigms. Spine (Phila Pa 1976) 2016; 41 Suppl 20:S218-S223. [PMID: 27488299 DOI: 10.1097/brs.0000000000001833] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY TYPE A review of the literature. OBJECTIVE The aim of this study was to discuss the evolution of molecular signatures and the history and development of targeted therapeutics in metastatic tumor types affecting the spinal column. SUMMARY OF BACKGROUND DATA Molecular characterization of metastatic spine tumors is expected to usher in a revolution in diagnostic and treatment paradigms. Molecular characterization will provide critical information that can be used for initial diagnosis, prognosticating the ideal treatment strategy, assessment of treatment efficacy, surveillance and monitoring recurrence, and predicting complications, clinical outcome, and overall survival in patients diagnosed with metastatic cancers to the spinal column. METHODS A review of the literature was performed focusing on illustrative examples of the role that molecular-based therapeutics have played in clinical outcomes for patients diagnosed with metastatic tumor types affecting the spinal column. RESULTS The impact of molecular therapeutics including receptor tyrosine kinases and immune checkpoint inhibitors and the ability of molecular signatures to provide prognostic information are discussed in metastatic breast cancer, lung cancer, prostate cancer, melanoma, and renal cell cancer affecting the spinal column. CONCLUSION For the providers who will ultimately counsel patients diagnosed with metastases to the spinal column, molecular advancements will radically alter the management/surgical paradigms utilized. Ultimately, the translation of these molecular advancements into routine clinical care will greatly improve the quality and quantity of life for patients diagnosed with spinal malignancies and provide better overall outcomes and counseling for treating physicians. LEVEL OF EVIDENCE N/A.
Collapse
|
33
|
Hu AX, Huang ZY, Liu P, Xiang T, Yan S, Zhang L. A new ten-gene risk fraction model serving as prognostic indicator for clinical outcome of multiple myeloma. Tumour Biol 2016; 37:10.1007/s13277-016-5449-4. [PMID: 27709552 DOI: 10.1007/s13277-016-5449-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 09/23/2016] [Indexed: 11/30/2022] Open
Abstract
Multiple myeloma (MM) is a kind of aggressive tumor prevalent with high heterogeneity. Abnormal expression of certain genes may lead to the occurrence and development of MM. Nowadays, it is not commonly seen in clinical research to predict the prognostic circumstances of patients with MM by multiple gene expression profiling method. Identification of potential genes in prognostic process could be beneficial for clinical management of MM. Therefore, we aimed to build a risk fraction model to screen out the prognostic indicator for clinical outcome of MM. Microarray data were downloaded from the Genome Expression Omnibus (GEO) datasets with accession numbers GSE24080 and GSE57317. A total of 279 samples were selected out randomly. Besides, a risk formula was constructed and verified in the dataset. Time-dependent receiver operating characteristic (ROC) curve was applied in evaluating the accurate prognostic conditions of patients. Finally, a ten genes model in the training dataset was found to be closely related to the survival condition of MM patients. Patients with MM were divided into two groups, high-risk and low-risk, by the expression of these ten genes, and significant statistical difference was found between the two groups. Furthermore, the result of multivariate cox regression and stratified analysis indicated that this model was independent of other clinical phenotypes. ROC curves also showed its feasibility to predict the survival status of MM patients. Our results demonstrated that the fraction risk model constructed by the selected ten genes could be used to predict survival status of multiple myeloma patients, which could also help in improvement of prognostic and therapeutic tool of MM.
Collapse
Affiliation(s)
- Ai-Xin Hu
- The Department of Orthopedic Surgery, People's Hospital of Three Gorges University, YiChang, Hubei Province, China
| | - Zhi-Yong Huang
- PuAi Institute, E Dong Healthcare Group, The Central Hospital of Huangshi, Huangshi, Hubei Province, China
| | - Ping Liu
- Department of Pathology, Shiyan Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Tian Xiang
- Department of Clinical Laboratory Center, Central Hospital of Enshi Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi, Hubei, China
| | - Shi Yan
- Department of Emergency, The Affiliated Huai'an Hospital of Xuzhou Medical College and The Second People's Hospital of Huai'an, Huai'an, China.
| | - Li Zhang
- Department of Hematology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China.
| |
Collapse
|
34
|
Geybels MS, Wright JL, Bibikova M, Klotzle B, Fan JB, Zhao S, Feng Z, Ostrander EA, Lin DW, Nelson PS, Stanford JL. Epigenetic signature of Gleason score and prostate cancer recurrence after radical prostatectomy. Clin Epigenetics 2016; 8:97. [PMID: 27651837 PMCID: PMC5024414 DOI: 10.1186/s13148-016-0260-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 08/24/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Identifying the subset of patients with clinically localized prostate cancer (PCa) at the highest risk of recurrence remains challenging, and better prognostic markers are needed. Gleason score is the best predictor of PCa aggressiveness and prognosis. In the present study, we generated an epigenetic signature based on high versus low Gleason score tumors and evaluated its ability to predict recurrence after radical prostatectomy. METHODS Genome-wide DNA methylation data from The Cancer Genome Atlas (TCGA; no. of patients = 333) and the elastic net method were used to generate an epigenetic signature by contrasting patients with high (8-10) versus low (≤6) Gleason score tumors. The signature was then tested in a cohort of 523 patients with clinically localized disease who had radical prostatectomy. Samples taken from the primary tumor were used for DNA methylation and mRNA expression profiling. Patients were followed for PCa recurrence on average for 8 years after diagnosis. RESULTS The epigenetic signature includes 52 differentially methylated CpG sites. In the testing cohort, the signature was associated with poorer recurrence-free survival (hazard ratio per 25 % increase = 1.78; 95 % confidence interval 1.48, 2.16). The signature significantly improved the area under the curve (AUC) for PCa recurrence compared to clinical-pathological parameters alone, particularly among patients diagnosed with Gleason score 7 tumors (0.64 vs. 0.76, P = 1.34E-4). Results were comparable for patients with Gleason 3 + 4 and those with 4 + 3 tumors. Gene Set Enrichment Analysis showed that higher levels of the signature were associated with increased expression of genes related to cell cycle proliferation and decreased expression of androgen-responsive genes. CONCLUSIONS This report shows evidence that DNA methylation patterns measured in prostate tumor cells are predictive of PCa aggressiveness. The epigenetic signature may have clinical utility to improve prognostication particularly in patients with intermediate Gleason score 7 tumors.
Collapse
Affiliation(s)
- Milan S Geybels
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109-1024 USA ; Department of Epidemiology, GROW School for Oncology and Developmental biology, Maastricht University, Maastricht, The Netherlands
| | - Jonathan L Wright
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109-1024 USA ; Department of Urology, University of Washington School of Medicine, Seattle, WA USA
| | | | | | - Jian-Bing Fan
- Illumina, Inc., San Diego, CA USA ; Current address: AnchorDx Corp., Guangzhou, 510300 People's Republic of China
| | - Shanshan Zhao
- Biostatistics & Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC USA
| | - Ziding Feng
- Department of Biostatistics, MD Anderson Cancer Center, Houston, TX USA
| | - Elaine A Ostrander
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, MD USA
| | - Daniel W Lin
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109-1024 USA ; Department of Urology, University of Washington School of Medicine, Seattle, WA USA
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA USA ; Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA USA ; Department of Medicine, University of Washington School of Medicine, Seattle, WA USA
| | - Janet L Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109-1024 USA ; Department of Epidemiology, University of Washington School of Public Health, Seattle, WA USA
| |
Collapse
|
35
|
You S, Knudsen BS, Erho N, Alshalalfa M, Takhar M, Al-Deen Ashab H, Davicioni E, Karnes RJ, Klein EA, Den RB, Ross AE, Schaeffer EM, Garraway IP, Kim J, Freeman MR. Integrated Classification of Prostate Cancer Reveals a Novel Luminal Subtype with Poor Outcome. Cancer Res 2016; 76:4948-58. [PMID: 27302169 PMCID: PMC5047668 DOI: 10.1158/0008-5472.can-16-0902] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/25/2016] [Indexed: 01/03/2023]
Abstract
Prostate cancer is a biologically heterogeneous disease with variable molecular alterations underlying cancer initiation and progression. Despite recent advances in understanding prostate cancer heterogeneity, better methods for classification of prostate cancer are still needed to improve prognostic accuracy and therapeutic outcomes. In this study, we computationally assembled a large virtual cohort (n = 1,321) of human prostate cancer transcriptome profiles from 38 distinct cohorts and, using pathway activation signatures of known relevance to prostate cancer, developed a novel classification system consisting of three distinct subtypes (named PCS1-3). We validated this subtyping scheme in 10 independent patient cohorts and 19 laboratory models of prostate cancer, including cell lines and genetically engineered mouse models. Analysis of subtype-specific gene expression patterns in independent datasets derived from luminal and basal cell models provides evidence that PCS1 and PCS2 tumors reflect luminal subtypes, while PCS3 represents a basal subtype. We show that PCS1 tumors progress more rapidly to metastatic disease in comparison with PCS2 or PCS3, including PSC1 tumors of low Gleason grade. To apply this finding clinically, we developed a 37-gene panel that accurately assigns individual tumors to one of the three PCS subtypes. This panel was also applied to circulating tumor cells (CTC) and provided evidence that PCS1 CTCs may reflect enzalutamide resistance. In summary, PCS subtyping may improve accuracy in predicting the likelihood of clinical progression and permit treatment stratification at early and late disease stages. Cancer Res; 76(17); 4948-58. ©2016 AACR.
Collapse
Affiliation(s)
- Sungyong You
- Division of Cancer Biology and Therapeutics, Departments of Surgery & Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Beatrice S Knudsen
- Division of Cancer Biology and Therapeutics, Departments of Surgery & Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Nicholas Erho
- GenomeDx Biosciences Inc., Vancouver, British Columbia, Canada
| | | | - Mandeep Takhar
- GenomeDx Biosciences Inc., Vancouver, British Columbia, Canada
| | | | - Elai Davicioni
- GenomeDx Biosciences Inc., Vancouver, British Columbia, Canada
| | | | - Eric A Klein
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, Ohio
| | - Robert B Den
- Department of Radiation Oncology, Jefferson Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Ashley E Ross
- Department of Urology, Northwestern University, Chicago, Illinois
| | | | - Isla P Garraway
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Jayoung Kim
- Division of Cancer Biology and Therapeutics, Departments of Surgery & Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Michael R Freeman
- Division of Cancer Biology and Therapeutics, Departments of Surgery & Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California.
| |
Collapse
|
36
|
Kim SH, Kim S, Joo J, Seo HK, Joung JY, Lee KH, Chung J. A retrospective study of predictive factors for unexpectedly prolonged or shortened progression-free survival and overall survival among patients with metastatic renal cell carcinoma who received first-line targeted therapy. BMC Cancer 2016; 16:577. [PMID: 27484254 PMCID: PMC4969738 DOI: 10.1186/s12885-016-2615-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 07/25/2016] [Indexed: 02/03/2023] Open
Abstract
Background To identify predictors of prolonged or shortened progression-free survival (PFS) and overall survival (OS) among patients with metastatic renal cell carcinoma (mRCC) who received first-line targeted therapies. Methods This retrospective study included 146 patients with mRCC who were treated during 2007–2015. These patients were divided into a group with the worst response (WG), an expected group (EG), and a group with the best response (BG), based on their PFS (≤3 monthsnths, 3–18 monthsnths, and >18 monthsnths, respectively) and OS (<1 year, 1–3 years, and >3 years, respectively). To identify significant predictive factors, the BG and WG were compared to the EG using the Memorial Sloan Kettering Cancer Center and Heng risk models. Results The overall PFS and OS were 9.3 months and 16.4 months, respectively. The median PFS for the WG (41.8 %), EG (45.9 %), and BG (12.3 %) were 2.7 months, 9.3 months, and 56.6 months, respectively, and the median OS for the WG (45.9 %), EG (35.6 %), and BG (18.5 %) were 5.5 months, 21.6 months, and 63.1 months, respectively; these outcomes were significantly different (p < 0.001). Nephrectomy (odds ratio [OR]: 7.15) was a significant predictor of PFS in the BG, and the significant predictors of OS in the BG were MSKCC intermediate risk (OR: 0.12), poor risk (OR: 0.04), and a disease-free interval of <1 year (OR: 0.23) (all, p < 0.05). Anemia (OR: 3.25) was a significant predictor of PFS in the WG, and the significant predictors of OS were age (OR: 1.05), anemia (OR: 4.13), lymphocytopenia (OR: 4.76), disease-free interval of <1 year (OR: 4.8), and synchronous metastasis (OR: 3.52) (all, p < 0.05). Conclusion We identified several significant predictors of unexpectedly good and poor response to first-line targeted therapy among patients with mRCC. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2615-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sung Han Kim
- Department of Urology, Center for Prostate Cancer, Hospital of National Cancer Center National Cancer Center, Goyang, Korea
| | - Sohee Kim
- Biometric Research Branch, Clinical Research Coordination Center, Hospital of National Cancer Center National Cancer Center, Goyang, Korea
| | - Jungnam Joo
- Biometric Research Branch, Clinical Research Coordination Center, Hospital of National Cancer Center National Cancer Center, Goyang, Korea
| | - Ho Kyung Seo
- Department of Urology, Center for Prostate Cancer, Hospital of National Cancer Center National Cancer Center, Goyang, Korea
| | - Jae Young Joung
- Department of Urology, Center for Prostate Cancer, Hospital of National Cancer Center National Cancer Center, Goyang, Korea
| | - Kang Hyun Lee
- Department of Urology, Center for Prostate Cancer, Hospital of National Cancer Center National Cancer Center, Goyang, Korea
| | - Jinsoo Chung
- Department of Urology, Center for Prostate Cancer, Hospital of National Cancer Center National Cancer Center, Goyang, Korea. .,Center for Prostate Cancer, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do, 410-769, Republic of Korea.
| |
Collapse
|
37
|
CYP17A1 Polymorphisms and Clinical Outcome of Castration-Resistant Prostate Cancer Patients Treated with Abiraterone. Int J Biol Markers 2016; 31:e264-9. [DOI: 10.5301/jbm.5000197] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2016] [Indexed: 11/20/2022]
Abstract
Background We evaluated the role of single nucleotide polymorphisms in the CYP17A1 gene for predicting clinical outcome in castration-resistant prostate cancer (CRPC) patients treated with abiraterone. Methods Sixty-four patients were genotyped for the selected polymorphisms (rs743572, rs10883783, rs17115100 and rs284849) in CYP17A1. We hypothesized that different genotypes could be associated with progression-free survival (PFS) and overall survival (OS). Results Statistical analyses highlighted no significant associations between these polymorphisms and clinical outcome. However, individuals with the most common TT genotype for rs10883783 had a 3 months’ longer PFS than individuals with the TA + AA genotype. Conclusions With the limitation of the retrospective study design and the small sample size, the analyzed polymorphisms do not seem to be correlated with clinical outcome of CRPC patients treated with abiraterone.
Collapse
|
38
|
Krstanoski Z, Vokac NK, Zagorac A, Pospihalj B, Munda M, Dzeroski S, Golouh R. TMPRSS2:ERG gene aberrations may provide insight into pT stage in prostate cancer. BMC Urol 2016; 16:35. [PMID: 27377958 PMCID: PMC4932765 DOI: 10.1186/s12894-016-0160-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 06/15/2016] [Indexed: 11/10/2022] Open
Abstract
Background TMPRSS2:ERG gene aberration may be a novel marker that improves risk stratification of prostate cancer before definitive cancer therapy, but studies have been inconclusive. Methods The study cohort consisted of 202 operable prostate cancer Slovenian patients who underwent laparoscopic radical prostatectomy. We retrospectively constructed tissue microarrays of their prostatic specimens for fluorescence in situ hybridization, with appropriate signals obtained in 148 patients for subsequent statistical analyses. Results The following genetic aberrations were found: TMPRSS2:ERG fusion, TMPRSS2 split (a non-ERG translocation) and ERG split (an ERG translocation without involvement of TMPRSS2). TMPRSS2:ERG gene fusion happened in 63 patients (42 %), TMPRSS2 split in 12 patients and ERG split in 8 patients. Association was tested between TMPRSS2:ERG gene fusion and several clinicopathological variables, i.e., pT stage, extended lymph node dissection status, and Gleason score, correcting for multiple comparisons. Only the association with pT stage was significant at p = 0.05: Of 62 patients with pT3 stage, 34 (55 %) had TMPRSS2:ERG gene fusion. In pT3 stage patients, stronger (but not significant) association between eLND status and TMPRSS2:ERG gene fusion was detected. We detected TMPRSS2:ERG gene fusion in 64 % of the pT3 stage patients where we did not perform an extended lymph node dissection. Conclusions Our results indicate that it is possible to predict pT3 stage at final histology from TMPRSS2:ERG gene fusion at initial core needle biopsy. FISH determination of TMPRSS2:ERG gene fusion may be particularly useful for patients scheduled to undergo a radical prostatectomy in order to improve oncological and functional results.
Collapse
Affiliation(s)
- Zoran Krstanoski
- Department of Urology, General Hospital Slovenj Gradec, Gosposvetska 1, 2380, Slovenj Gradec, Slovenia.
| | - Nadja Kokalj Vokac
- Laboratory of Medical Genetics, University Medical Centre Maribor, Maribor, Slovenia
| | - Andreja Zagorac
- Laboratory of Medical Genetics, University Medical Centre Maribor, Maribor, Slovenia
| | - Boris Pospihalj
- Division of Gynecology, Department of Gynecological Pathology and Cytology, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Miha Munda
- Institute for Anatomy, Histology and Embryology, Medical Faculty, University of Maribor, Maribor, Slovenia
| | | | - Rastko Golouh
- Institute of Pathology, Medical Faculty University of Maribor, Maribor, Slovenia
| |
Collapse
|
39
|
Hu BR, Fairey AS, Madhav A, Yang D, Li M, Groshen S, Stephens C, Kim PH, Virk N, Wang L, Martin SE, Erho N, Davicioni E, Jenkins RB, Den RB, Xu T, Xu Y, Gill IS, Quinn DI, Goldkorn A. AXIN2 expression predicts prostate cancer recurrence and regulates invasion and tumor growth. Prostate 2016; 76:597-608. [PMID: 26771938 PMCID: PMC7455032 DOI: 10.1002/pros.23151] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 12/31/2015] [Indexed: 01/16/2023]
Abstract
BACKGROUND Treatment of prostate cancer (PCa) may be improved by identifying biological mechanisms of tumor growth that directly impact clinical disease progression. We investigated whether genes associated with a highly tumorigenic, drug resistant, progenitor phenotype impact PCa biology and recurrence. METHODS Radical prostatectomy (RP) specimens (±disease recurrence, N = 276) were analyzed by qRT-PCR to quantify expression of genes associated with self-renewal, drug resistance, and tumorigenicity in prior studies. Associations between gene expression and PCa recurrence were confirmed by bootstrap internal validation and by external validation in independent cohorts (total N = 675) and in silico. siRNA knockdown and lentiviral overexpression were used to determine the effect of gene expression on PCa invasion, proliferation, and tumor growth. RESULTS Four candidate genes were differentially expressed in PCa recurrence. Of these, low AXIN2 expression was internally validated in the discovery cohort. Validation in external cohorts and in silico demonstrated that low AXIN2 was independently associated with more aggressive PCa, biochemical recurrence, and metastasis-free survival after RP. Functionally, siRNA-mediated depletion of AXIN2 significantly increased invasiveness, proliferation, and tumor growth. Conversely, ectopic overexpression of AXIN2 significantly reduced invasiveness, proliferation, and tumor growth. CONCLUSIONS Low AXIN2 expression was associated with PCa recurrence after RP in our test population as well as in external validation cohorts, and its expression levels in PCa cells significantly impacted invasiveness, proliferation, and tumor growth. Given these novel roles, further study of AXIN2 in PCa may yield promising new predictive and therapeutic strategies.
Collapse
Affiliation(s)
- Brian R. Hu
- USC Institute of Urology, Keck Medical Center of USC and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Adrian S. Fairey
- USC Institute of Urology, Keck Medical Center of USC and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Anisha Madhav
- Division of Medical Oncology, Department of Medicine, University of Southern California Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, Los Angeles, California
| | - Dongyun Yang
- Department of Preventive Medicine, Keck Medical Center of USC, University of Southern California, Los Angeles, California
| | - Meng Li
- Health Sciences Bioinformatics Core, USC Keck School of Medicine, Los Angeles, California
| | - Susan Groshen
- Department of Preventive Medicine, Keck Medical Center of USC, University of Southern California, Los Angeles, California
| | | | - Philip H. Kim
- USC Institute of Urology, Keck Medical Center of USC and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Navneet Virk
- Division of Medical Oncology, Department of Medicine, University of Southern California Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, Los Angeles, California
| | - Lina Wang
- Department of Pathology, Keck Medical Center of USC, University of Southern California, Los Angeles, California
| | - Sue Ellen Martin
- Department of Pathology, Keck Medical Center of USC, University of Southern California, Los Angeles, California
| | | | | | - Robert B. Jenkins
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Robert B. Den
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Tong Xu
- Division of Medical Oncology, Department of Medicine, University of Southern California Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, Los Angeles, California
| | - Yucheng Xu
- Division of Medical Oncology, Department of Medicine, University of Southern California Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, Los Angeles, California
| | - Inderbir S. Gill
- USC Institute of Urology, Keck Medical Center of USC and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - David I. Quinn
- Division of Medical Oncology, Department of Medicine, University of Southern California Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, Los Angeles, California
| | - Amir Goldkorn
- Division of Medical Oncology, Department of Medicine, University of Southern California Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, Los Angeles, California
| |
Collapse
|
40
|
Okihara K, Ochiai A, Kamoi K, Fujizuka Y, Miki T, Ito K. Comprehensive assessment for novel prostate cancer markers in the prostate-specific antigen era: focusing on Asians and Asian countries. Int J Urol 2016; 22:334-41. [PMID: 25827049 DOI: 10.1111/iju.12701] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 11/17/2014] [Accepted: 12/03/2014] [Indexed: 11/27/2022]
Abstract
We reviewed the current evidence for three novel prostate tumor markers (PCA3, TMPRSS2:ERG and proPSA) that have been recently reported predominantly in Western countries. We focus our attention on Asian men in both clinical and basic research studies. There have been no reports on the clinical usefulness of these three markers for Asians living in Western countries. In Asian countries, evidence for the clinical usefulness of PCA3 and proPSA-related indices including Prostate Health Index is being accumulated, mainly in Japan. The process for how a novel marker is approved in the clinical setting is also discussed.
Collapse
Affiliation(s)
- Koji Okihara
- Department of Urology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | | | | | | | | |
Collapse
|
41
|
Mohsenzadegan M, Shekarabi M, Madjd Z, Asgari M, Abolhasani M, Tajik N, Farajollahi MM. Study of NGEP expression pattern in cancerous tissues provides novel insights into prognostic marker in prostate cancer. Biomark Med 2016; 9:391-401. [PMID: 25808443 DOI: 10.2217/bmm.14.106] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
AIM The aim of this study was to produce a novel polyclonal antibody against extracellular domain of NGEP protein and explore its role in prognosis of prostate cancer. MATERIALS & METHODS Polyclonal antibodies against two peptides (NGEP-p1 and NGEP-p2) derived extracellular domains of NGEP were prepared and the intensity and distribution of NGEP expression analyzed in large series of prostate tissue specimens. RESULTS We found a significant inverse correlation between NGEP expression and prognostic features such as Gleason score, pathologic tumor stage and prostate-specific antigen level using anti-NGEP-p2 antibody. CONCLUSION The results indicate that the high level of expression could be associated with good prognosis in prostate cancer. However, additional studies are required to evaluate NGEP as an independent prognostic factor for prostate carcinoma.
Collapse
Affiliation(s)
- Monireh Mohsenzadegan
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | | | |
Collapse
|
42
|
Peng Z, Andersson K, Lindholm J, Dethlefsen O, Pramana S, Pawitan Y, Nistér M, Nilsson S, Li C. Improving the Prediction of Prostate Cancer Overall Survival by Supplementing Readily Available Clinical Data with Gene Expression Levels of IGFBP3 and F3 in Formalin-Fixed Paraffin Embedded Core Needle Biopsy Material. PLoS One 2016; 11:e0145545. [PMID: 26731648 PMCID: PMC4701463 DOI: 10.1371/journal.pone.0145545] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 12/04/2015] [Indexed: 11/28/2022] Open
Abstract
Background A previously reported expression signature of three genes (IGFBP3, F3 and VGLL3) was shown to have potential prognostic value in estimating overall and cancer-specific survivals at diagnosis of prostate cancer in a pilot cohort study using freshly frozen Fine Needle Aspiration (FNA) samples. Methods We carried out a new cohort study with 241 prostate cancer patients diagnosed from 2004–2007 with a follow-up exceeding 6 years in order to verify the prognostic value of gene expression signature in formalin fixed paraffin embedded (FFPE) prostate core needle biopsy tissue samples. The cohort consisted of four patient groups with different survival times and death causes. A four multiplex one-step RT-qPCR test kit, designed and optimized for measuring the expression signature in FFPE core needle biopsy samples, was used. In archive FFPE biopsy samples the expression differences of two genes (IGFBP3 and F3) were measured. The survival time predictions using the current clinical parameters only, such as age at diagnosis, Gleason score, PSA value and tumor stage, and clinical parameters supplemented with the expression levels of IGFBP3 and F3, were compared. Results When combined with currently used clinical parameters, the gene expression levels of IGFBP3 and F3 are improving the prediction of survival time as compared to using clinical parameters alone. Conclusion The assessment of IGFBP3 and F3 gene expression levels in FFPE prostate cancer tissue would provide an improved survival prediction for prostate cancer patients at the time of diagnosis.
Collapse
Affiliation(s)
- Zhuochun Peng
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Chundsell Medicals AB, Stockholm, Sweden
| | - Karl Andersson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Ridgeview Instruments AB, Uppsala, Sweden
| | - Johan Lindholm
- Clinical Pathology/Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Olga Dethlefsen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Setia Pramana
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Yudi Pawitan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Monica Nistér
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Clinical Pathology/Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Sten Nilsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Clinical Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Chunde Li
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Clinical Oncology, Karolinska University Hospital, Stockholm, Sweden
- Chundsell Medicals AB, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
43
|
Core 2 β-1, 6-N-acetylglucosaminyltransferase-1 expression in prostate biopsy specimen is an indicator of prostate cancer aggressiveness. Biochem Biophys Res Commun 2016; 470:150-156. [PMID: 26768364 DOI: 10.1016/j.bbrc.2016.01.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 01/04/2016] [Indexed: 11/22/2022]
Abstract
INTRODUCTION To avoid over-treatment of early stage prostate cancer (PCa), predictive biomarkers for PCa aggressiveness which can be obtained during pre-treatment evaluation are essential. Core 2 β-1, 6-N-acetylglucosaminyl-transferase-1 (GCNT1) is a key enzyme that forms core 2 branched O-glycans, the expression of which is associated with aggressive potential of prostate cancer. We examined whether GCNT1 expression in prostate biopsy specimen can predict cancer recurrence after radical prostatectomy for the patients with with PCa. We then investigated molecular background for aggressive malignant potential mediated by GCNT1 expression. METHODS Paraffin-embedded PCa biopsy specimens were immunohisto-chemically tested for GCNT1 expression using an anti-GCNT1 monoclonal antibody. We also examined the role of GCNT1 in PCa progression using cell lines which express high or low levels of GCNT1. RESULTS GCNT1 expression correlated with D' Amico's recurrence risk classification. The GCNT1-positive rate in organ confined PCa was significantly lower than that in PCa with extra-prostatic extension. GCNT1-negative tumors were associated with significantly better prostate-specific antigen (PSA)-free survival compared with GCNT1-positive tumors. Multivariate analysis revealed that GCNT1 expression status was an independent risk factor for PSA recurrence after radical prostatectomy. Subsequent basic study revealed that GCNT1-over-expressing cells produced a significantly larger amount of growth factors when co-cultured with prostate stromal cells compared with GCNT1-knocked down cells and formed larger tumors. CONCLUSIONS GCNT1 expression in prostate biopsy specimen is a significant and independent predictor of recurrence after radical prostatectomy, which can be used in pre-treatment decision making for the patient. Further validation study is necessary to establish clinical implication of GCNT1 in management of PCa.
Collapse
|
44
|
Abstract
Men of African origin are disproportionately affected by prostate cancer: prostate cancer incidence is highest among men of African origin in the USA, prostate cancer mortality is highest among men of African origin in the Caribbean, and tumour stage and grade at diagnosis are highest among men in sub-Saharan Africa. Socioeconomic, educational, cultural, and genetic factors, as well as variations in care delivery and treatment selection, contribute to this cancer disparity. Emerging data on single-nucleotide-polymorphism patterns, epigenetic changes, and variations in fusion-gene products among men of African origin add to the understanding of genetic differences underlying this disease. On the diagnosis of prostate cancer, when all treatment options are available, men of African origin are more likely to choose radiation therapy or to receive no definitive treatment than white men. Among men of African origin undergoing surgery, increased rates of biochemical recurrence have been identified. Understanding differences in the cancer-survivorship experience and quality-of-life outcomes among men of African origin are critical to appropriately counsel patients and improve cultural sensitivity. Efforts to curtail prostate cancer screening will likely affect men of African origin disproportionately and widen the racial disparity of disease.
Collapse
|
45
|
Loss of heterozygosity for chromosomal regions 15q14-21.1, 17q21.31, and 13q12.3-13.1 and its relevance for prostate cancer. Med Oncol 2015; 32:246. [PMID: 26433958 PMCID: PMC4592700 DOI: 10.1007/s12032-015-0691-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 09/24/2015] [Indexed: 01/07/2023]
Abstract
Although prostate cancer is one of the most common cancers in men, the genetic defects underlying its pathogenesis remain poorly understood. DNA damage repair mechanisms have been implicated in human cancer. Accumulating evidence indicates that the fidelity of the response to DNA double-strand breaks is critical for maintaining genome integrity. RAD51 is a central player in double-strand break repair via homologous recombination, and its alterations may confer and increase the risk of cancer. RAD51 functioning depends on the indirect or direct interactions with BRCA1 and BRCA2. To evaluate the contribution of RAD51 to sporadic prostate cancer, loss of heterozygosity (LOH) for chromosomal region 15q14-21.1 (RAD51locus) was determined and compared to LOH in 17q21.31 (BRCA1 locus) and 13q12.3-13.1 (BRCA2 region). DNA was isolated from prostate biopsies and matched peripheral blood of 50 patients. The regions 15q14-21.1, 17q21.31, and 13q12.3-13.1 were examined using microsatellite markers on chromosome 15 (D15S118, D15S214, D15S1006), chromosome 17 (D17S855, D17S1323), and chromosome 13 (D13S260, D13S290), respectively. The LOH in tumors was analyzed by PCR with fluorescently labeled primers and an ABI PRISM 377 DNA Sequencer. Allele sizing was determined by GeneScan version 3.1.2 and Genotyper version 2.5 software (Applied Biosystems, USA). LOH was identified in 57.5, 23, and 40 % for chromosomal regions 15q14-21.1, 17q21.31, and 13q12.3-13.1, respectively. Twenty-six percent of studied cases manifested LOH for at least one marker in 15q14-21.1 exclusively. A significant correlation was found between LOH for studied region and PSAD (prostate-specific antigen density). The findings suggest that RAD51 may be considered as a prostate cancer susceptibility gene.
Collapse
|
46
|
Kojima Y, Yoneyama T, Hatakeyama S, Mikami J, Sato T, Mori K, Hashimoto Y, Koie T, Ohyama C, Fukuda M, Tobisawa Y. Detection of Core2 β-1,6-N-Acetylglucosaminyltransferase in Post-Digital Rectal Examination Urine Is a Reliable Indicator for Extracapsular Extension of Prostate Cancer. PLoS One 2015; 10:e0138520. [PMID: 26390303 PMCID: PMC4577128 DOI: 10.1371/journal.pone.0138520] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 09/01/2015] [Indexed: 11/19/2022] Open
Abstract
To identify appropriate candidates for aggressive treatment such as radical prostatectomy or radiation therapy of localized prostate cancer (PCa), novel predictive biomarkers of PCa aggressiveness are essential. Core2 β-1,6-N-acetylglucosaminyltransferase-1 (GCNT1) is a key enzyme that forms core 2-branched O-glycans. Its expression is associated with the progression of several cancers. We established a mouse IgG monoclonal antibody (mAb) against GCNT1 and examined the relationship of GCNT1 expression to the clinicopathological status of PCa. Paraffin-embedded PCa specimens were analyzed by immunohistochemistry for GCNT1 expression using a newly established mouse anti-GCNT1 mAb by ourselves. GCNT1-positive tumor showed significantly higher Gleason score and larger tumor volume. The number of GCNT1-positive cases was significantly lower in cases of organ-confined disease than in cases of extracapsular extension. GCNT1-negative tumors were associated with significantly better prostate-specific antigen (PSA)-free survival compared with GCNT1-positive tumors. Multivariate analysis revealed that detection of GCNT1 expression was an independent risk factor for PSA recurrence. We established new methods for GCNT1 detection from PCa specimens. Immunoblotting was used to examine post-digital rectal examination (DRE) urine from PCa patients. Over 90% of GCNT1-positive PCa patients with high concentrations of PSA showed extracapsular extension. In conclusion, GCNT1 expression closely associates with the aggressive potential of PCa. Further research aims to develop GCNT1 detection in post-DRE urine as a marker for PCa aggressiveness.
Collapse
Affiliation(s)
- Yuta Kojima
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Tohru Yoneyama
- Department of Advanced Transplant and Regenerative Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Shingo Hatakeyama
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Jotaro Mikami
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Tendo Sato
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kazuyuki Mori
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yasuhiro Hashimoto
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Takuya Koie
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Chikara Ohyama
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
- Department of Advanced Transplant and Regenerative Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Minoru Fukuda
- Glycobiology Unit, Tumor Microenvironment Program, Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, United States of America
| | - Yuki Tobisawa
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
- * E-mail:
| |
Collapse
|
47
|
Gentles AJ, Bratman SV, Lee LJ, Harris JP, Feng W, Nair RV, Shultz DB, Nair VS, Hoang CD, West RB, Plevritis SK, Alizadeh AA, Diehn M. Integrating Tumor and Stromal Gene Expression Signatures With Clinical Indices for Survival Stratification of Early-Stage Non-Small Cell Lung Cancer. J Natl Cancer Inst 2015; 107:djv211. [PMID: 26286589 DOI: 10.1093/jnci/djv211] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 07/07/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Accurate survival stratification in early-stage non-small cell lung cancer (NSCLC) could inform the use of adjuvant therapy. We developed a clinically implementable mortality risk score incorporating distinct tumor microenvironmental gene expression signatures and clinical variables. METHODS Gene expression profiles from 1106 nonsquamous NSCLCs were used for generation and internal validation of a nine-gene molecular prognostic index (MPI). A quantitative polymerase chain reaction (qPCR) assay was developed and validated on an independent cohort of formalin-fixed paraffin-embedded (FFPE) tissues (n = 98). A prognostic score using clinical variables was generated using Surveillance, Epidemiology, and End Results data and combined with the MPI. All statistical tests for survival were two-sided. RESULTS The MPI stratified stage I patients into prognostic categories in three microarray and one FFPE qPCR validation cohorts (HR = 2.99, 95% CI = 1.55 to 5.76, P < .001 in stage IA patients of the largest microarray validation cohort; HR = 3.95, 95% CI = 1.24 to 12.64, P = .01 in stage IA of the qPCR cohort). Prognostic genes were expressed in distinct tumor cell subpopulations, and genes implicated in proliferation and stem cells portended poor outcomes, while genes involved in normal lung differentiation and immune infiltration were associated with superior survival. Integrating the MPI with clinical variables conferred greatest prognostic power (HR = 3.43, 95% CI = 2.18 to 5.39, P < .001 in stage I patients of the largest microarray cohort; HR = 3.99, 95% CI = 1.67 to 9.56, P < .001 in stage I patients of the qPCR cohort). Finally, the MPI was prognostic irrespective of somatic alterations in EGFR, KRAS, TP53, and ALK. CONCLUSION The MPI incorporates genes expressed in the tumor and its microenvironment and can be implemented clinically using qPCR assays on FFPE tissues. A composite model integrating the MPI with clinical variables provides the most accurate risk stratification.
Collapse
Affiliation(s)
- Andrew J Gentles
- Department of Radiology (AJG, JPH, RVN, SKP), Department of Radiation Oncology (SVB, DBS, MD), Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine (LJL, WF, MD), Department of Medicine Division of Pulmonary and Critical Care Medicine (VSN), Department of Cardiothoracic Surgery Division of Thoracic Surgery (CDH), Department of Pathology (RBW), and Department of Medicine Division of Oncology (AAA), Stanford University, Stanford, CA
| | - Scott V Bratman
- Department of Radiology (AJG, JPH, RVN, SKP), Department of Radiation Oncology (SVB, DBS, MD), Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine (LJL, WF, MD), Department of Medicine Division of Pulmonary and Critical Care Medicine (VSN), Department of Cardiothoracic Surgery Division of Thoracic Surgery (CDH), Department of Pathology (RBW), and Department of Medicine Division of Oncology (AAA), Stanford University, Stanford, CA
| | - Luke J Lee
- Department of Radiology (AJG, JPH, RVN, SKP), Department of Radiation Oncology (SVB, DBS, MD), Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine (LJL, WF, MD), Department of Medicine Division of Pulmonary and Critical Care Medicine (VSN), Department of Cardiothoracic Surgery Division of Thoracic Surgery (CDH), Department of Pathology (RBW), and Department of Medicine Division of Oncology (AAA), Stanford University, Stanford, CA
| | - Jeremy P Harris
- Department of Radiology (AJG, JPH, RVN, SKP), Department of Radiation Oncology (SVB, DBS, MD), Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine (LJL, WF, MD), Department of Medicine Division of Pulmonary and Critical Care Medicine (VSN), Department of Cardiothoracic Surgery Division of Thoracic Surgery (CDH), Department of Pathology (RBW), and Department of Medicine Division of Oncology (AAA), Stanford University, Stanford, CA
| | - Weiguo Feng
- Department of Radiology (AJG, JPH, RVN, SKP), Department of Radiation Oncology (SVB, DBS, MD), Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine (LJL, WF, MD), Department of Medicine Division of Pulmonary and Critical Care Medicine (VSN), Department of Cardiothoracic Surgery Division of Thoracic Surgery (CDH), Department of Pathology (RBW), and Department of Medicine Division of Oncology (AAA), Stanford University, Stanford, CA
| | - Ramesh V Nair
- Department of Radiology (AJG, JPH, RVN, SKP), Department of Radiation Oncology (SVB, DBS, MD), Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine (LJL, WF, MD), Department of Medicine Division of Pulmonary and Critical Care Medicine (VSN), Department of Cardiothoracic Surgery Division of Thoracic Surgery (CDH), Department of Pathology (RBW), and Department of Medicine Division of Oncology (AAA), Stanford University, Stanford, CA
| | - David B Shultz
- Department of Radiology (AJG, JPH, RVN, SKP), Department of Radiation Oncology (SVB, DBS, MD), Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine (LJL, WF, MD), Department of Medicine Division of Pulmonary and Critical Care Medicine (VSN), Department of Cardiothoracic Surgery Division of Thoracic Surgery (CDH), Department of Pathology (RBW), and Department of Medicine Division of Oncology (AAA), Stanford University, Stanford, CA
| | - Viswam S Nair
- Department of Radiology (AJG, JPH, RVN, SKP), Department of Radiation Oncology (SVB, DBS, MD), Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine (LJL, WF, MD), Department of Medicine Division of Pulmonary and Critical Care Medicine (VSN), Department of Cardiothoracic Surgery Division of Thoracic Surgery (CDH), Department of Pathology (RBW), and Department of Medicine Division of Oncology (AAA), Stanford University, Stanford, CA
| | - Chuong D Hoang
- Department of Radiology (AJG, JPH, RVN, SKP), Department of Radiation Oncology (SVB, DBS, MD), Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine (LJL, WF, MD), Department of Medicine Division of Pulmonary and Critical Care Medicine (VSN), Department of Cardiothoracic Surgery Division of Thoracic Surgery (CDH), Department of Pathology (RBW), and Department of Medicine Division of Oncology (AAA), Stanford University, Stanford, CA
| | - Robert B West
- Department of Radiology (AJG, JPH, RVN, SKP), Department of Radiation Oncology (SVB, DBS, MD), Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine (LJL, WF, MD), Department of Medicine Division of Pulmonary and Critical Care Medicine (VSN), Department of Cardiothoracic Surgery Division of Thoracic Surgery (CDH), Department of Pathology (RBW), and Department of Medicine Division of Oncology (AAA), Stanford University, Stanford, CA
| | - Sylvia K Plevritis
- Department of Radiology (AJG, JPH, RVN, SKP), Department of Radiation Oncology (SVB, DBS, MD), Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine (LJL, WF, MD), Department of Medicine Division of Pulmonary and Critical Care Medicine (VSN), Department of Cardiothoracic Surgery Division of Thoracic Surgery (CDH), Department of Pathology (RBW), and Department of Medicine Division of Oncology (AAA), Stanford University, Stanford, CA.
| | - Ash A Alizadeh
- Department of Radiology (AJG, JPH, RVN, SKP), Department of Radiation Oncology (SVB, DBS, MD), Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine (LJL, WF, MD), Department of Medicine Division of Pulmonary and Critical Care Medicine (VSN), Department of Cardiothoracic Surgery Division of Thoracic Surgery (CDH), Department of Pathology (RBW), and Department of Medicine Division of Oncology (AAA), Stanford University, Stanford, CA.
| | - Maximilian Diehn
- Department of Radiology (AJG, JPH, RVN, SKP), Department of Radiation Oncology (SVB, DBS, MD), Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine (LJL, WF, MD), Department of Medicine Division of Pulmonary and Critical Care Medicine (VSN), Department of Cardiothoracic Surgery Division of Thoracic Surgery (CDH), Department of Pathology (RBW), and Department of Medicine Division of Oncology (AAA), Stanford University, Stanford, CA.
| |
Collapse
|
48
|
Prognostic role of genetic biomarkers in clinical progression of prostate cancer. Exp Mol Med 2015; 47:e176. [PMID: 26251261 PMCID: PMC4558485 DOI: 10.1038/emm.2015.43] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 03/17/2015] [Accepted: 03/19/2015] [Indexed: 01/01/2023] Open
Abstract
The aim of this study was to analyze the use of 12 single-nucleotide polymorphisms in genes ELAC2, RNASEL and MSR1 as biomarkers for prostate cancer (PCa) detection and progression, as well as perform a genetic classification of high-risk patients. A cohort of 451 men (235 patients and 216 controls) was studied. We calculated means of regression analysis using clinical values (stage, prostate-specific antigen, Gleason score and progression) in patients and controls at the basal stage and after a follow-up of 72 months. Significantly different allele frequencies between patients and controls were observed for rs1904577 and rs918 (MSR1 gene) and for rs17552022 and rs5030739 (ELAC2). We found evidence of increased risk for PCa in rs486907 and rs2127565 in variants AA and CC, respectively. In addition, rs627928 (TT-GT), rs486907 (AG) and rs3747531 (CG-CC) were associated with low tumor aggressiveness. Some had a weak linkage, such as rs1904577 and rs2127565, rs4792311 and rs17552022, and rs1904577 and rs918. Our study provides the proof-of-principle that some of the genetic variants (such as rs486907, rs627928 and rs2127565) in genes RNASEL, MSR1 and ELAC2 can be used as predictors of aggressiveness and progression of PCa. In the future, clinical use of these biomarkers, in combination with current ones, could potentially reduce the rate of unnecessary biopsies and specific treatments.
Collapse
|
49
|
Stattin P, Vickers AJ, Sjoberg DD, Johansson R, Granfors T, Johansson M, Pettersson K, Scardino PT, Hallmans G, Lilja H. Improving the Specificity of Screening for Lethal Prostate Cancer Using Prostate-specific Antigen and a Panel of Kallikrein Markers: A Nested Case-Control Study. Eur Urol 2015; 68:207-13. [PMID: 25682340 PMCID: PMC4496315 DOI: 10.1016/j.eururo.2015.01.009] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/09/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND A disadvantage of prostate-specific antigen (PSA) for the early detection of prostate cancer (PCa) is that many men must be screened, biopsied, and diagnosed to prevent one death. OBJECTIVE To increase the specificity of screening for lethal PCa at an early stage. DESIGN, SETTING, AND PARTICIPANTS We conducted a case-control study nested within a population-based cohort. PSA and three additional kallikreins were measured in cryopreserved blood from a population-based cohort in Västerbotten, Sweden. Of 40379 men providing blood at ages 40, 50, and 60 yr from 1986 to 2009, 12542 men were followed for >15 yr. From this cohort, the Swedish Cancer Registry identified 1423 incident PCa cases, 235 with distant metastasis. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Risk of distant metastasis for different PSA levels and a prespecified statistical model based on the four kallikrein markers. RESULTS AND LIMITATIONS Most metastatic cases occurred in men with PSA in the top quartile at age 50 yr (69%) or 60 yr (74%), whereas 20-yr risk of metastasis for men with PSA below median was low (≤0.6%). Among men with PSA >2 ng/ml, a prespecified model based on four kallikrein markers significantly enhanced the prediction of metastasis compared with PSA alone. About half of all men with PSA >2 ng/ml were defined as low risk by this model and had a ≤1% 15-yr risk of metastasis. CONCLUSIONS Screening at ages 50-60 yr should focus on men with PSA in the top quartile. A marker panel can aid biopsy decision making. PATIENT SUMMARY For men in their fifties, screening should focus on those in the top 10% to 25% of PSA values because the majority of subsequent cases of distant metastasis are found among these men. Testing of four kallikrein markers in men with an elevated PSA could aid biopsy decision making.
Collapse
Affiliation(s)
- Pär Stattin
- Department of Surgical and Perioperative Sciences, Urology and Andrology, Umeå University, Umeå, Sweden
| | - Andrew J Vickers
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel D Sjoberg
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert Johansson
- Regional Cancer Centre, Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | | | - Mattias Johansson
- Section of Genetics, The International Agency for Research on Cancer, Lyon, France
| | - Kim Pettersson
- Division of Biotechnology, University of Turku, Turku, Finland
| | - Peter T Scardino
- Department of Surgery (Urology), Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Göran Hallmans
- Department of Public Health and Clinical Medicine, Nutritional Research, Umeå University, Umeå, Sweden
| | - Hans Lilja
- Department of Surgery (Urology), Memorial Sloan Kettering Cancer Center, New York, NY, USA; Departments of Laboratory Medicine and Medicine (Genitourinary Oncology), Memorial Sloan Kettering Cancer Center, New York, NY, USA; Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Headington, Oxford, UK; Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Recent advances in sequencing technologies have allowed for the identification of genetic variants within germline DNA that can explain a significant portion of the genetic underpinnings of prostate cancer. Despite evidence suggesting that these genetic variants can be used for improved risk stratification, they have not yet been routinely incorporated into routine clinical practice. This review highlights their potential utility in prostate cancer screening. RECENT FINDINGS There are now almost 100 genetic variants, called single nucleotide polymorphisms (SNPs) that have been recently found to be associated with the risk of developing prostate cancer. In addition, some of these prostate cancer risk SNPs have also been found to influence prostate specific antigen (PSA) expression levels and potentially aggressive disease. SUMMARY Incorporation of panels of prostate cancer risk SNPs into clinical practice offers potential to provide improvements in patient selection for prostate cancer screening; PSA interpretation (e.g. by correcting for the presence of SNPs that influence PSA expression levels; decision for biopsy (using prostate cancer risk SNPs); and possibly the decision for treatment. A proposed clinical algorithm incorporating these prostate cancer risk SNPs is discussed.
Collapse
|