1
|
Fennessy FM, Maier SE. Quantitative diffusion MRI in prostate cancer: Image quality, what we can measure and how it improves clinical assessment. Eur J Radiol 2023; 167:111066. [PMID: 37651828 PMCID: PMC10623580 DOI: 10.1016/j.ejrad.2023.111066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/19/2023] [Accepted: 08/24/2023] [Indexed: 09/02/2023]
Abstract
Diffusion-weighted imaging is a dependable method for detection of clinically significant prostate cancer. In prostate tissue, there are several compartments that can be distinguished from each other, based on different water diffusion decay signals observed. Alterations in cell architecture, such as a relative increase in tumor infiltration and decrease in stroma, will influence the observed diffusion signal in a voxel due to impeded random motion of water molecules. The amount of restricted diffusion can be assessed quantitatively by measuring the apparent diffusion coefficient (ADC) value. This is traditionally calculated using a monoexponential decay formula represented by the slope of a line produced between the logarithm of signal intensity decay plotted against selected b-values. However, the choice and number of b-values and their distribution, has a significant effect on the measured ADC values. There have been many models that attempt to use higher-order functions to better describe the observed diffusion signal decay, requiring an increased number and range of b-values. While ADC can probe heterogeneity on a macroscopic level, there is a need to optimize advanced diffusion techniques to better interrogate prostate tissue microstructure. This could be of benefit in clinical challenges such as identifying sparse tumors in normal prostate tissue or better defining tumor margins. This paper reviews the principles of diffusion MRI and novel higher order diffusion signal analysis techniques to improve the detection of prostate cancer.
Collapse
Affiliation(s)
- Fiona M Fennessy
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| | - Stephan E Maier
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States; Department of Radiology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
2
|
Albisinni S, Sarkis J, Diamand R, De Nunzio C. Prebiopsy 68Ga-PSMA PET imaging: can we improve the current diagnostic pathway for prostate cancer? Prostate Cancer Prostatic Dis 2023; 26:47-49. [PMID: 36085498 DOI: 10.1038/s41391-022-00593-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/09/2022] [Accepted: 08/31/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Simone Albisinni
- Department of Urology, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium.
- Department of Urology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
- Urology Unit, Department of Surgical Sciences, Tor Vergata University Hospital, University of Rome Tor Vergata, Rome, Italy.
| | - Julien Sarkis
- Department of Urology, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Romain Diamand
- Department of Urology, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
- Department of Urology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Cosimo De Nunzio
- Department of Urology, Ospedale Sant'Andrea, Sapienza University, Roma, Italy
| |
Collapse
|
3
|
Robinson H, Roberts MJ, Gardiner RA, Hill MM. Extracellular vesicles for precision medicine in prostate cancer - Is it ready for clinical translation? Semin Cancer Biol 2023; 89:18-29. [PMID: 36681206 DOI: 10.1016/j.semcancer.2023.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023]
Abstract
Biofluid-based biomarker tests hold great promise for precision medicine in prostate cancer (PCa) clinical practice. Extracellular vesicles (EV) are established as intercellular messengers in cancer development with EV cargos, including protein and nucleic acids, having the potential to serve as biofluid-based biomarkers. Recent clinical studies have begun to evaluate EV-based biomarkers for PCa diagnosis, prognosis, and disease/therapy resistance monitoring. Promising results have led to PCa EV biomarker validation studies which are currently underway with the next challenge being translation to robust clinical assays. However, EV research studies generally use low throughput EV isolation methods and costly molecular profiling technologies that are not suitable for clinical assays. Here, we consider the technical hurdles in translating EV biomarker research findings into precise and cost-effective clinical biomarker assays. Novel microfluidic devices coupling EV extraction with sensitive antibody-based biomarker detection are already being explored for point-of-care applications for rapid provision in personalised medicine approaches.
Collapse
Affiliation(s)
- Harley Robinson
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, Queensland, Australia.
| | - Matthew J Roberts
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Brisbane, Queensland, Australia; Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Robert A Gardiner
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Brisbane, Queensland, Australia; Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Michelle M Hill
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, Queensland, Australia; UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Brisbane, Queensland, Australia.
| |
Collapse
|
4
|
Eickelschulte S, Riediger AL, Angeles AK, Janke F, Duensing S, Sültmann H, Görtz M. Biomarkers for the Detection and Risk Stratification of Aggressive Prostate Cancer. Cancers (Basel) 2022; 14:cancers14246094. [PMID: 36551580 PMCID: PMC9777028 DOI: 10.3390/cancers14246094] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Current strategies for the clinical management of prostate cancer are inadequate for a precise risk stratification between indolent and aggressive tumors. Recently developed tissue-based molecular biomarkers have refined the risk assessment of the disease. The characterization of tissue biopsy components and subsequent identification of relevant tissue-based molecular alterations have the potential to improve the clinical decision making and patient outcomes. However, tissue biopsies are invasive and spatially restricted due to tumor heterogeneity. Therefore, there is an urgent need for complementary diagnostic and prognostic options. Liquid biopsy approaches are minimally invasive with potential utility for the early detection, risk stratification, and monitoring of tumors. In this review, we focus on tissue and liquid biopsy biomarkers for early diagnosis and risk stratification of prostate cancer, including modifications on the genomic, epigenomic, transcriptomic, and proteomic levels. High-risk molecular alterations combined with orthogonal clinical parameters can improve the identification of aggressive tumors and increase patient survival.
Collapse
Affiliation(s)
- Samaneh Eickelschulte
- Junior Clinical Cooperation Unit, Multiparametric Methods for Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Urology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Anja Lisa Riediger
- Junior Clinical Cooperation Unit, Multiparametric Methods for Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Urology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Arlou Kristina Angeles
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Florian Janke
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Stefan Duensing
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Holger Sültmann
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Magdalena Görtz
- Junior Clinical Cooperation Unit, Multiparametric Methods for Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Urology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-6221-42-2603
| |
Collapse
|
5
|
Okubo Y, Yamamoto Y, Terao H, Suzuki T, Koizumi M, Yoshioka E, Washimi K, Sato S, Yokose T, Kishida T, Miyagi Y. Significance of non-standardized magnetic resonance imaging abnormalities and subsequent targeted prostate cancer biopsy for pathologists: A retrospective observational study. Pathol Res Pract 2022; 240:154188. [DOI: 10.1016/j.prp.2022.154188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 11/05/2022]
|
6
|
Pidsley R, Lam D, Qu W, Peters TJ, Luu P, Korbie D, Stirzaker C, Daly RJ, Stricker P, Kench JG, Horvath LG, Clark SJ. Comprehensive methylome sequencing reveals prognostic epigenetic biomarkers for prostate cancer mortality. Clin Transl Med 2022; 12:e1030. [PMID: 36178085 PMCID: PMC9523674 DOI: 10.1002/ctm2.1030] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Prostate cancer is a clinically heterogeneous disease with a subset of patients rapidly progressing to lethal-metastatic prostate cancer. Current clinicopathological measures are imperfect predictors of disease progression. Epigenetic changes are amongst the earliest molecular changes in tumourigenesis. To find new prognostic biomarkers to enable earlier intervention and improved outcomes, we performed methylome sequencing of DNA from patients with localised prostate cancer and long-term clinical follow-up. METHODS We used whole-genome bisulphite sequencing (WGBS) to comprehensively map and compare DNA methylation of radical prostatectomy tissue between patients with lethal disease (n = 7) and non-lethal (n = 8) disease (median follow-up 19.5 years). Validation of differentially methylated regions (DMRs) was performed in an independent cohort (n = 185, median follow-up 15 years) using targeted multiplex bisulphite sequencing of candidate regions. Survival was assessed via univariable and multivariable analyses including clinicopathological measures (log-rank and Cox regression models). RESULTS WGBS data analysis identified cancer-specific methylation patterns including CpG island hypermethylation, and hypomethylation of repetitive elements, with increasing disease risk. We identified 1420 DMRs associated with prostate cancer-specific mortality (PCSM), which showed enrichment for gene sets downregulated in prostate cancer and de novo methylated in cancer. Through comparison with public prostate cancer datasets, we refined the DMRs to develop an 18-gene prognostic panel. Applying this panel to an independent cohort, we found significant associations between PCSM and hypermethylation at EPHB3, PARP6, TBX1, MARCH6 and a regulatory element within CACNA2D4. Strikingly in a multivariable model, inclusion of CACNA2D4 methylation was a better predictor of PCSM versus grade alone (Harrell's C-index: 0.779 vs. 0.684). CONCLUSIONS Our study provides detailed methylome maps of non-lethal and lethal prostate cancer and identifies novel genic regions that distinguish these patient groups. Inclusion of our DNA methylation biomarkers with existing clinicopathological measures improves prognostic models of prostate cancer mortality, and holds promise for clinical application.
Collapse
Affiliation(s)
- Ruth Pidsley
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia,School of Clinical MedicineSt Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW SydneySydneyNew South WalesAustralia
| | - Dilys Lam
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia,Present address:
School of Molecular Sciences, The University of Western Australia, Crawley, Western Australia 6009, Australia,Present address:
Harry Perkins Institute of Medical Research, Nedlands, Western Australia 6009, Australia
| | - Wenjia Qu
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia
| | - Timothy J. Peters
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia,School of Clinical MedicineSt Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW SydneySydneyNew South WalesAustralia
| | - Phuc‐Loi Luu
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia,School of Clinical MedicineSt Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW SydneySydneyNew South WalesAustralia
| | - Darren Korbie
- Centre for Personalised NanomedicineAustralian Institute for Bioengineering and NanotechnologyThe University of QueenslandSt. LuciaQueenslandAustralia
| | - Clare Stirzaker
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia,School of Clinical MedicineSt Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW SydneySydneyNew South WalesAustralia
| | - Roger J. Daly
- Cancer Research Program and Department of Biochemistry and Molecular BiologyBiomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
| | - Phillip Stricker
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia,School of Clinical MedicineSt Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW SydneySydneyNew South WalesAustralia,Department of UrologySt. Vincent's Prostate Cancer CentreSydneyNew South WalesAustralia
| | - James G. Kench
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia,Department of Tissue PathologyNSW Health PathologyRoyal Prince Alfred HospitalCamperdownSydneyNew South WalesAustralia
| | - Lisa G. Horvath
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia,School of Clinical MedicineSt Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW SydneySydneyNew South WalesAustralia,Chris O'Brien Lifehouse, CamperdownSydneyNew South WalesAustralia,University of SydneySydneyNew South WalesAustralia
| | - Susan J. Clark
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia,School of Clinical MedicineSt Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW SydneySydneyNew South WalesAustralia
| |
Collapse
|
7
|
Volatilomics: An Emerging and Promising Avenue for the Detection of Potential Prostate Cancer Biomarkers. Cancers (Basel) 2022; 14:cancers14163982. [PMID: 36010975 PMCID: PMC9406416 DOI: 10.3390/cancers14163982] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 12/20/2022] Open
Abstract
Simple Summary The lack of highly specific and sensitive biomarkers for the early detection of prostate cancer (PCa) is a major barrier to its management. Volatilomics emerged as a non-invasive, simple, inexpensive, and easy-to-use approach for cancer screening, characterization of disease progression, and follow-up of the treatment’s success. We provide a brief overview of the potential of volatile organic metabolites (VOMs) for the establishment of PCa biomarkers from non-invasive matrices. Endogenous VOMs have been investigated as potential biomarkers since changes in these VOMs can be characteristic of specific disease processes. Recent studies have shown that the conjugation of the prostate-specific antigen (PSA) screening with other methodologies, such as risk calculators, biomarkers, and imaging tests, can attenuate overdiagnosis and under-detection issues. This means that the combination of volatilomics with other methodologies could be extremely valuable for the differentiation of clinical phenotypes in a group of patients, providing more personalized treatments. Abstract Despite the spectacular advances in molecular medicine, including genomics, proteomics, transcriptomics, lipidomics, and personalized medicine, supported by the discovery of the human genome, prostate cancer (PCa) remains the most frequent malignant tumor and a leading cause of oncological death in men. New methods for prognostic, diagnostic, and therapy evaluation are mainly based on the combination of imaging techniques with other methodologies, such as gene or protein profiling, aimed at improving PCa management and surveillance. However, the lack of highly specific and sensitive biomarkers for its early detection is a major hurdle to this goal. Apart from classical biomarkers, the study of endogenous volatile organic metabolites (VOMs) biosynthesized by different metabolic pathways and found in several biofluids is emerging as an innovative, efficient, accessible, and non-invasive approach to establish the volatilomic biosignature of PCa patients, unravelling potential biomarkers. This review provides a brief overview of the challenges of PCa screening methods and emergent biomarkers. We also focus on the potential of volatilomics for the establishment of PCa biomarkers from non-invasive matrices.
Collapse
|
8
|
Prostate Cancer Radiogenomics-From Imaging to Molecular Characterization. Int J Mol Sci 2021; 22:ijms22189971. [PMID: 34576134 PMCID: PMC8465891 DOI: 10.3390/ijms22189971] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/06/2021] [Accepted: 09/10/2021] [Indexed: 12/24/2022] Open
Abstract
Radiomics and genomics represent two of the most promising fields of cancer research, designed to improve the risk stratification and disease management of patients with prostate cancer (PCa). Radiomics involves a conversion of imaging derivate quantitative features using manual or automated algorithms, enhancing existing data through mathematical analysis. This could increase the clinical value in PCa management. To extract features from imaging methods such as magnetic resonance imaging (MRI), the empiric nature of the analysis using machine learning and artificial intelligence could help make the best clinical decisions. Genomics information can be explained or decoded by radiomics. The development of methodologies can create more-efficient predictive models and can better characterize the molecular features of PCa. Additionally, the identification of new imaging biomarkers can overcome the known heterogeneity of PCa, by non-invasive radiological assessment of the whole specific organ. In the future, the validation of recent findings, in large, randomized cohorts of PCa patients, can establish the role of radiogenomics. Briefly, we aimed to review the current literature of highly quantitative and qualitative results from well-designed studies for the diagnoses, treatment, and follow-up of prostate cancer, based on radiomics, genomics and radiogenomics research.
Collapse
|
9
|
Basourakos SP, Tzeng M, Lewicki PJ, Patel K, Al Hussein Al Awamlh B, Venkat S, Shoag JE, Gorin MA, Barbieri CE, Hu JC. Tissue-Based Biomarkers for the Risk Stratification of Men With Clinically Localized Prostate Cancer. Front Oncol 2021; 11:676716. [PMID: 34123846 PMCID: PMC8193839 DOI: 10.3389/fonc.2021.676716] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/21/2021] [Indexed: 01/09/2023] Open
Abstract
Risk stratification of men with clinically localized prostate cancer has historically relied on basic clinicopathologic parameters such as prostate specific antigen level, grade group, and clinical stage. However, prostate cancer often behaves in ways that cannot be accurately predicted by these parameters. Thus, recent efforts have focused on developing tissue-based genomic tests that provide greater insights into the risk of a given patient's disease. Multiple tests are now commercially available and provide additional prognostic information at various stages of the care pathway for prostate cancer. Indeed, early evidence suggests that these assays may have a significant impact on patient and physician decision-making. However, the impact of these tests on oncologic outcomes remains less clear. In this review, we highlight recent advances in the use of tissue-based biomarkers in the treatment of prostate cancer and identify the existing evidence supporting their clinical use.
Collapse
Affiliation(s)
- Spyridon P. Basourakos
- Department of Urology, New York-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - Michael Tzeng
- Department of Urology, New York-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - Patrick J. Lewicki
- Department of Urology, New York-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - Krishnan Patel
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD, United States
| | | | - Siv Venkat
- Department of Urology, New York-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - Jonathan E. Shoag
- Department of Urology, New York-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
- Department of Urology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Michael A. Gorin
- Department of Urology, University of Pittsburg School of Medicine, Pittsburgh, PA, United States
- Urology Associates and UPMC Western Maryland, Cumberland, MD, United States
| | - Christopher E. Barbieri
- Department of Urology, New York-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - Jim C. Hu
- Department of Urology, New York-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
10
|
Turnham DJ, Bullock N, Dass MS, Staffurth JN, Pearson HB. The PTEN Conundrum: How to Target PTEN-Deficient Prostate Cancer. Cells 2020; 9:E2342. [PMID: 33105713 PMCID: PMC7690430 DOI: 10.3390/cells9112342] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/17/2020] [Accepted: 10/20/2020] [Indexed: 12/17/2022] Open
Abstract
Loss of the tumor suppressor phosphatase and tensin homologue deleted on chromosome 10 (PTEN), which negatively regulates the PI3K-AKT-mTOR pathway, is strongly linked to advanced prostate cancer progression and poor clinical outcome. Accordingly, several therapeutic approaches are currently being explored to combat PTEN-deficient tumors. These include classical inhibition of the PI3K-AKT-mTOR signaling network, as well as new approaches that restore PTEN function, or target PTEN regulation of chromosome stability, DNA damage repair and the tumor microenvironment. While targeting PTEN-deficient prostate cancer remains a clinical challenge, new advances in the field of precision medicine indicate that PTEN loss provides a valuable biomarker to stratify prostate cancer patients for treatments, which may improve overall outcome. Here, we discuss the clinical implications of PTEN loss in the management of prostate cancer and review recent therapeutic advances in targeting PTEN-deficient prostate cancer. Deepening our understanding of how PTEN loss contributes to prostate cancer growth and therapeutic resistance will inform the design of future clinical studies and precision-medicine strategies that will ultimately improve patient care.
Collapse
Affiliation(s)
- Daniel J. Turnham
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
| | - Nicholas Bullock
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK;
| | - Manisha S. Dass
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
| | - John N. Staffurth
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK;
| | - Helen B. Pearson
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
| |
Collapse
|
11
|
Lam D, Clark S, Stirzaker C, Pidsley R. Advances in Prognostic Methylation Biomarkers for Prostate Cancer. Cancers (Basel) 2020; 12:E2993. [PMID: 33076494 PMCID: PMC7602626 DOI: 10.3390/cancers12102993] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 12/24/2022] Open
Abstract
There is a major clinical need for accurate biomarkers for prostate cancer prognosis, to better inform treatment strategies and disease monitoring. Current clinically recognised prognostic factors, including prostate-specific antigen (PSA) levels, lack sensitivity and specificity in distinguishing aggressive from indolent disease, particularly in patients with localised intermediate grade prostate cancer. There has therefore been a major focus on identifying molecular biomarkers that can add prognostic value to existing markers, including investigation of DNA methylation, which has a known role in tumorigenesis. In this review, we will provide a comprehensive overview of the current state of DNA methylation biomarker studies in prostate cancer prognosis, and highlight the advances that have been made in this field. We cover the numerous studies into well-established candidate genes, and explore the technological transition that has enabled hypothesis-free genome-wide studies and the subsequent discovery of novel prognostic genes.
Collapse
Affiliation(s)
- Dilys Lam
- Epigenetics Research Laboratory, Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; (D.L.); (S.C.); (C.S.)
| | - Susan Clark
- Epigenetics Research Laboratory, Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; (D.L.); (S.C.); (C.S.)
- St. Vincent’s Clinical School, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Clare Stirzaker
- Epigenetics Research Laboratory, Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; (D.L.); (S.C.); (C.S.)
- St. Vincent’s Clinical School, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Ruth Pidsley
- Epigenetics Research Laboratory, Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; (D.L.); (S.C.); (C.S.)
- St. Vincent’s Clinical School, University of New South Wales, Sydney, New South Wales 2010, Australia
| |
Collapse
|
12
|
Toth R, Schiffmann H, Hube-Magg C, Büscheck F, Höflmayer D, Weidemann S, Lebok P, Fraune C, Minner S, Schlomm T, Sauter G, Plass C, Assenov Y, Simon R, Meiners J, Gerhäuser C. Random forest-based modelling to detect biomarkers for prostate cancer progression. Clin Epigenetics 2019; 11:148. [PMID: 31640781 PMCID: PMC6805338 DOI: 10.1186/s13148-019-0736-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/03/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The clinical course of prostate cancer (PCa) is highly variable, demanding an individualized approach to therapy. Overtreatment of indolent PCa cases, which likely do not progress to aggressive stages, may be associated with severe side effects and considerable costs. These could be avoided by utilizing robust prognostic markers to guide treatment decisions. RESULTS We present a random forest-based classification model to predict aggressive behaviour of prostate cancer. DNA methylation changes between PCa cases with good or poor prognosis (discovery cohort with n = 70) were used as input. DNA was extracted from formalin-fixed tumour tissue, and genome-wide DNA methylation differences between both groups were assessed using Illumina HumanMethylation450 arrays. For the random forest-based modelling, the discovery cohort was randomly split into a training (80%) and a test set (20%). Our methylation-based classifier demonstrated excellent performance in discriminating prognosis subgroups in the test set (Kaplan-Meier survival analyses with log-rank p value < 0.0001). The area under the receiver operating characteristic curve (AUC) for the sensitivity analysis was 95%. Using the ICGC cohort of early- and late-onset prostate cancer (n = 222) and the TCGA PRAD cohort (n = 477) for external validation, AUCs for sensitivity analyses were 77.1% and 68.7%, respectively. Cancer progression-related DNA hypomethylation was frequently located in 'partially methylated domains' (PMDs)-large-scale genomic areas with progressive loss of DNA methylation linked to mitotic cell division. We selected several candidate genes with differential methylation in gene promoter regions for additional validation at the protein expression level by immunohistochemistry in > 12,000 tissue micro-arrayed PCa cases. Loss of ZIC2 protein expression was associated with poor prognosis and correlated with significantly shorter time to biochemical recurrence. The prognostic value of ZIC2 proved to be independent from established clinicopathological variables including Gleason grade, tumour stage, nodal stage and prostate-specific-antigen. CONCLUSIONS Our results highlight the prognostic relevance of methylation loss in PMD regions, as well as of several candidate genes not previously associated with PCa progression. Our robust and externally validated PCa classification model either directly or via protein expression analyses of the identified top-ranked candidate genes will support the clinical management of prostate cancer.
Collapse
Affiliation(s)
- Reka Toth
- Cancer Epigenomics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Heiko Schiffmann
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Claudia Hube-Magg
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Franziska Büscheck
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Doris Höflmayer
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Sören Weidemann
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Patrick Lebok
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Christoph Fraune
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Sarah Minner
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Thorsten Schlomm
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.,Department of Urology, Charité Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Guido Sauter
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Christoph Plass
- Cancer Epigenomics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
| | - Yassen Assenov
- Cancer Epigenomics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Ronald Simon
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Jan Meiners
- General, Visceral and Thoracic Surgery Department and Clinic, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Clarissa Gerhäuser
- Cancer Epigenomics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.
| |
Collapse
|
13
|
Singh S, Gill AA, Nlooto M, Karpoormath R. Prostate cancer biomarkers detection using nanoparticles based electrochemical biosensors. Biosens Bioelectron 2019; 137:213-221. [DOI: 10.1016/j.bios.2019.03.065] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/08/2019] [Accepted: 03/18/2019] [Indexed: 02/07/2023]
|
14
|
Vermassen T, De Bruyne S, Himpe J, Lumen N, Callewaert N, Rottey S, Delanghe J. N-Linked Glycosylation and Near-Infrared Spectroscopy in the Diagnosis of Prostate Cancer. Int J Mol Sci 2019; 20:ijms20071592. [PMID: 30934974 PMCID: PMC6479798 DOI: 10.3390/ijms20071592] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 03/21/2019] [Accepted: 03/23/2019] [Indexed: 01/07/2023] Open
Abstract
Background: Performing a prostate biopsy is the most robust and reliable way to diagnose prostate cancer (PCa), and to determine the disease grading. As little to no biochemical markers for prostate tissue exist, we explored the possibilities of tissue N-glycosylation and near-infrared spectroscopy (NIR) in PCa diagnosis. Methods: Tissue specimens from 100 patients (benign prostate hyperplasia (BPH), n = 50; and PCa, n = 50) were obtained. The fresh-frozen tissue was dispersed and a tissue N-glycosylation profile was determined. Consequently, the formalin-fixed paraffin-embedded slides were analyzed using NIR spectroscopy. A comparison was made between the benign and malignant tissue, and between the various Gleason scores. Results: A difference was observed for the tissue of N-glycosylation between the benign and malignant tissue. These differences were located in the fycosylation ratios and the total amount of bi- and tetra-antennary structures (all p < 0.0001). These differences were also present between various Gleason scores. In addition, the NIR spectra revealed changes between the benign and malignant tissue in several regions. Moreover, spectral ranges of 1055–1065 nm and 1450–1460 nm were significantly different between the Gleason scores (p = 0.0042 and p = 0.0195). Conclusions: We have demonstrated biochemical changes in the N-glycan profile of prostate tissue, which allows for the distinction between malignant and benign tissue, as well as between various Gleason scores. These changes can be correlated to the changes observed in the NIR spectra. This could possibly further improve the histological assessment of PCa diagnosis, although further method validation is needed.
Collapse
Affiliation(s)
- Tijl Vermassen
- Department of Medical Oncology, Ghent University Hospital, 9000 Ghent, Belgium.
| | - Sander De Bruyne
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, 9000 Ghent, Belgium.
| | - Jonas Himpe
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, 9000 Ghent, Belgium.
| | - Nicolaas Lumen
- Department of Urology, Ghent University Hospital, 9000 Ghent, Belgium.
| | - Nico Callewaert
- Unit for Medical Biotechnology, Inflammation Research Center, VIB⁻Ghent University, 9052 Zwijnaarde, Belgium.
| | - Sylvie Rottey
- Department of Medical Oncology, Ghent University Hospital, 9000 Ghent, Belgium.
| | - Joris Delanghe
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, 9000 Ghent, Belgium.
- Department of Clinical Chemistry, Ghent University Hospital, 9000 Ghent, Belgium.
| |
Collapse
|
15
|
Turiák L, Ozohanics O, Tóth G, Ács A, Révész Á, Vékey K, Telekes A, Drahos L. High sensitivity proteomics of prostate cancer tissue microarrays to discriminate between healthy and cancerous tissue. J Proteomics 2018; 197:82-91. [PMID: 30439472 DOI: 10.1016/j.jprot.2018.11.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 08/28/2018] [Accepted: 11/09/2018] [Indexed: 01/14/2023]
Abstract
Biopsies, in the form of tissue microarrays (TMAs) were studied to identify anomalies indicative of prostate cancer at the proteome level. TMAs offer a valuable source of well-characterized biological material. However, because of the small tissue sample size method development was essential to provide the sensitivity and reliability necessary for the analysis. Surface digestion of TMA cores was followed by peptide extraction and shotgun proteomics analysis. About 5 times better sensitivity was achieved by the optimized surface digestion compared to bulk digestion of the same TMA spot and it allowed the identification of over 500 proteins from individual prostate TMA cores. Label-free quantitation showed that biological variability among all samples was about 3 times larger than the technical reproducibility. We have identified 189 proteins which showed statistically significant changes (t-test p-value <.05) in abundance between healthy and cancerous tissue samples. The proteomic profile changed according to cancer grade, but did not show a correlation with cancer stage. Results of this pilot study were further evaluated using bioinformatics tools, identifying various protein pathways affected by prostate cancer progression indicating the usefulness of studying TMA cores to identify quantitative changes in tissue proteomics. SIGNIFICANCE: Detailed proteomics analysis of TMAs presents a good alternative for tissue analysis. Here we present a novel method, based on tissue surface digestion and nano-LC-MS measurements, which is capable of identifying and quantifying over 500 proteins from a 1.5 mm diameter tissue section. We compared healthy and cancerous prostate tissue samples, and tissues with various grades and stages of cancer. Tissue proteomics clearly distinguished healthy and cancerous samples, furthermore the results correlated well with cancer grade, but not with cancer stage. Over 100 proteins showed statistically significant abundance changes (t-test p-value <.05) between various groups. This was sufficient for a meaningful bioinformatics evaluation; showing e.g. increased abundance of proteins in cancer in the KEGG ribosome pathway, GO mRNA splicing via spliceosome, and chromatin assembly biological processes. The results highlight the feasibility of the developed method for future large-scale tissue proteomics studies using commercially available TMAs.
Collapse
Affiliation(s)
- Lilla Turiák
- MS Proteomics Research Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, H-1117 Budapest, Hungary.
| | - Oliver Ozohanics
- MS Proteomics Research Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, H-1117 Budapest, Hungary
| | - Gábor Tóth
- MS Proteomics Research Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, H-1117 Budapest, Hungary; Budapest University of Technology and Economics, Faculty of Chemical Technology and Biotechnology, Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - András Ács
- MS Proteomics Research Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, H-1117 Budapest, Hungary; Semmelweis University, Ph.D. School of Pharmaceutical Sciences, Üllői út 26, H-1085 Budapest, Hungary
| | - Ágnes Révész
- MS Proteomics Research Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, H-1117 Budapest, Hungary
| | - Károly Vékey
- MS Proteomics Research Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, H-1117 Budapest, Hungary
| | - András Telekes
- Div. Sect. of Geriatrics, 2nd Department of Internal Medicine, Semmelweis University, Halmi utca 20-22, H-1115 Budapest, Hungary; Dept. of Oncology, St Lazarus County Hospital, Füleki út 54-56, H-1117, Salgótarján, Hungary
| | - László Drahos
- MS Proteomics Research Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, H-1117 Budapest, Hungary
| |
Collapse
|
16
|
Salami SS, Hovelson DH, Kaplan JB, Mathieu R, Udager AM, Curci NE, Lee M, Plouffe KR, de la Vega LL, Susani M, Rioux-Leclercq N, Spratt DE, Morgan TM, Davenport MS, Chinnaiyan AM, Cyrta J, Rubin MA, Shariat SF, Tomlins SA, Palapattu GS. Transcriptomic heterogeneity in multifocal prostate cancer. JCI Insight 2018; 3:123468. [PMID: 30385730 DOI: 10.1172/jci.insight.123468] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/27/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Commercial gene expression assays are guiding clinical decision making in patients with prostate cancer, particularly when considering active surveillance. Given heterogeneity and multifocality of primary prostate cancer, such assays should ideally be robust to the coexistence of unsampled higher grade disease elsewhere in the prostate in order to have clinical utility. Herein, we comprehensively evaluated transcriptomic profiles of primary multifocal prostate cancer to assess robustness to clinically relevant multifocality. METHODS We designed a comprehensive, multiplexed targeted RNA-sequencing assay capable of assessing multiple transcriptional classes and deriving commercially available prognostic signatures, including the Myriad Prolaris Cell Cycle Progression score, the Oncotype DX Genomic Prostate Score, and the GenomeDX Decipher Genomic Classifier. We applied this assay to a retrospective, multi-institutional cohort of 156 prostate cancer samples. Derived commercial biomarker scores for 120 informative primary prostate cancer samples from 44 cases were determined and compared. RESULTS Derived expression scores were positively correlated with tumor grade (rS = 0.53-0.73; all P < 0.001), both within the same case and across the entire cohort. In cases of extreme grade-discordant multifocality (co-occurrence of grade group 1 [GG1] and ≥GG4 foci], gene expression scores were significantly lower in low- (GG1) versus high-grade (≥GG4) foci (all P < 0.001). No significant differences in expression scores, however, were observed between GG1 foci from prostates with and without coexisting higher grade cancer (all P > 0.05). CONCLUSIONS Multifocal, low-grade and high-grade prostate cancer foci exhibit distinct prognostic expression signatures. These findings demonstrate that prognostic RNA expression assays performed on low-grade prostate cancer biopsy tissue may not provide meaningful information on the presence of coexisting unsampled aggressive disease. FUNDING Prostate Cancer Foundation, National Institutes of Health (U01 CA214170, R01 CA183857, University of Michigan Prostate Specialized Program of Research Excellence [S.P.O.R.E.] P50 CA186786-05, Weill Cornell Medicine S.P.O.R.E. P50 CA211024-01A1), Men of Michigan Prostate Cancer Research Fund, University of Michigan Comprehensive Cancer Center core grant (2-P30-CA-046592-24), A. Alfred Taubman Biomedical Research Institute, and Department of Defense.
Collapse
Affiliation(s)
- Simpa S Salami
- Department of Urology, Michigan Medicine, Ann Arbor, Michigan, USA.,University of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA
| | - Daniel H Hovelson
- Department of Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Jeremy B Kaplan
- Department of Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Romain Mathieu
- Department of Urology, Medical University Vienna, Vienna, Austria.,Department of Urology, Rennes University Hospital, Rennes, France
| | - Aaron M Udager
- Department of Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Nicole E Curci
- Department of Radiology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Matthew Lee
- Department of Urology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Komal R Plouffe
- Department of Pathology, Michigan Medicine, Ann Arbor, Michigan, USA
| | | | - Martin Susani
- Department of Pathology, Medical University Vienna, Vienna, Austria
| | | | - Daniel E Spratt
- University of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA.,Department of Radiation Oncology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Todd M Morgan
- Department of Urology, Michigan Medicine, Ann Arbor, Michigan, USA.,University of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA
| | - Matthew S Davenport
- Department of Urology, Michigan Medicine, Ann Arbor, Michigan, USA.,Department of Radiology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Arul M Chinnaiyan
- Department of Urology, Michigan Medicine, Ann Arbor, Michigan, USA.,University of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA.,Department of Pathology, Michigan Medicine, Ann Arbor, Michigan, USA.,Michigan Center for Translational Pathology, Ann Arbor, Michigan, USA
| | - Joanna Cyrta
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, USA
| | - Mark A Rubin
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, USA.,Department of BioMedical Research, University of Bern, Bern, Switzerland
| | | | - Scott A Tomlins
- Department of Urology, Michigan Medicine, Ann Arbor, Michigan, USA.,University of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA.,Department of Pathology, Michigan Medicine, Ann Arbor, Michigan, USA.,Michigan Center for Translational Pathology, Ann Arbor, Michigan, USA
| | - Ganesh S Palapattu
- Department of Urology, Michigan Medicine, Ann Arbor, Michigan, USA.,University of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA.,Department of Urology, Medical University Vienna, Vienna, Austria
| |
Collapse
|
17
|
Soodana-Prakash N, Stoyanova R, Bhat A, Velasquez MC, Kineish OE, Pollack A, Parekh DJ, Punnen S. Entering an era of radiogenomics in prostate cancer risk stratification. Transl Androl Urol 2018; 7:S443-S452. [PMID: 30363524 PMCID: PMC6178317 DOI: 10.21037/tau.2018.07.04] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Radiogenomics is a field that amalgamates data from genomics and imaging techniques in order to derive clinically meaningful trends. In this article, we discuss the importance of prostate cancer risk classification and how data derived from genomic testing and multi-parametric magnetic resonance imaging (mpMRI) can be integrated into clinical decision-making processes with a focus on active surveillance (AS). Finally, we describe an ongoing prospective trial (Miami MAST trial) which incorporates imaging (mpMRI) and radiomics data in patients who are on AS for prostate cancer.
Collapse
Affiliation(s)
| | - Radka Stoyanova
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA.,Department of Radiation Oncology, University of Miami, Miami, FL, USA
| | - Abhishek Bhat
- Department of Urology, University of Miami, Miami, FL, USA
| | | | - Omer E Kineish
- Department of Urology, University of Miami, Miami, FL, USA
| | - Alan Pollack
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA.,Department of Radiation Oncology, University of Miami, Miami, FL, USA
| | - Dipen J Parekh
- Department of Urology, University of Miami, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Sanoj Punnen
- Department of Urology, University of Miami, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| |
Collapse
|
18
|
Angeles AK, Bauer S, Ratz L, Klauck SM, Sültmann H. Genome-Based Classification and Therapy of Prostate Cancer. Diagnostics (Basel) 2018; 8:E62. [PMID: 30200539 PMCID: PMC6164491 DOI: 10.3390/diagnostics8030062] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 12/19/2022] Open
Abstract
In the past decade, multi-national and multi-center efforts were launched to sequence prostate cancer genomes, transcriptomes, and epigenomes with the aim of discovering the molecular underpinnings of tumorigenesis, cancer progression, and therapy resistance. Multiple biological markers and pathways have been discovered to be tumor drivers, and a molecular classification of prostate cancer is emerging. Here, we highlight crucial findings of these genome-sequencing projects in localized and advanced disease. We recapitulate the utility and limitations of current clinical practices to diagnosis, prognosis, and therapy, and we provide examples of insights generated by the molecular profiling of tumors. Novel treatment concepts based on these molecular alterations are currently being addressed in clinical trials and will lead to an enhanced implementation of precision medicine strategies.
Collapse
Affiliation(s)
- Arlou Kristina Angeles
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, Heidelberg D-69120, Germany.
| | - Simone Bauer
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, Heidelberg D-69120, Germany.
| | - Leonie Ratz
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, Heidelberg D-69120, Germany.
| | - Sabine M Klauck
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, Heidelberg D-69120, Germany.
| | - Holger Sültmann
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, Heidelberg D-69120, Germany.
| |
Collapse
|