1
|
Adorno Febles VR, Hao Y, Ahsan A, Wu J, Qian Y, Zhong H, Loeb S, Makarov DV, Lepor H, Wysock J, Taneja SS, Huang WC, Becker DJ, Balar AV, Melamed J, Deng FM, Ren Q, Kufe D, Wong KK, Adeegbe DO, Deng J, Wise DR. Single-cell analysis of localized prostate cancer patients links high Gleason score with an immunosuppressive profile. Prostate 2023; 83:840-849. [PMID: 36988342 DOI: 10.1002/pros.24524] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 02/18/2023] [Accepted: 03/02/2023] [Indexed: 03/30/2023]
Abstract
BACKGROUND Evading immune surveillance is a hallmark for the development of multiple cancer types. Whether immune evasion contributes to the pathogenesis of high-grade prostate cancer (HGPCa) remains an area of active inquiry. METHODS Through single-cell RNA sequencing and multicolor flow cytometry of freshly isolated prostatectomy specimens and matched peripheral blood, we aimed to characterize the tumor immune microenvironment (TME) of localized prostate cancer (PCa), including HGPCa and low-grade prostate cancer (LGPCa). RESULTS HGPCa are highly infiltrated by exhausted CD8+ T cells, myeloid cells, and regulatory T cells (TRegs). These HGPCa-infiltrating CD8+ T cells expressed high levels of exhaustion markers including TIM3, TOX, TCF7, PD-1, CTLA4, TIGIT, and CXCL13. By contrast, a high ratio of activated CD8+ effector T cells relative to TRegs and myeloid cells infiltrate the TME of LGPCa. HGPCa CD8+ tumor-infiltrating lymphocytes (TILs) expressed more androgen receptor and prostate-specific membran antigen yet less prostate-specific antigen than the LGPCa CD8+ TILs. The PCa TME was infiltrated by macrophages but these did not clearly cluster by M1 and M2 markers. CONCLUSIONS Our study reveals a suppressive TME with high levels of CD8+ T cell exhaustion in localized PCa, a finding enriched in HGPCa relative to LGPCa. These studies suggest a possible link between the clinical-pathologic risk of PCa and the associated TME. Our results have implications for our understanding of the immunologic mechanisms of PCa pathogenesis and the implementation of immunotherapy for localized PCa.
Collapse
Affiliation(s)
- Victor R Adorno Febles
- Department of Medicine, Laura & Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
- Department of Medicine, Manhattan Campus, VA NY Harbor Health Care System, New York, New York, USA
| | - Yuan Hao
- Applied Bioinformatics Laboratories, New York University Langone Health, New York, New York, USA
| | - Aarif Ahsan
- Department of Medicine, Laura & Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Jiansheng Wu
- Department of Medicine, Laura & Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Yingzhi Qian
- Department of Population Health, NYU Langone Health, New York, New York, USA
| | - Hua Zhong
- Department of Population Health, NYU Langone Health, New York, New York, USA
| | - Stacy Loeb
- Department of Urology, New York University School of Medicine, New York, New York, USA
- Department of Urology, Manhattan Campus, VA NY Harbor Health Care System, New York, New York, USA
| | - Danil V Makarov
- Department of Urology, New York University School of Medicine, New York, New York, USA
- Department of Urology, Manhattan Campus, VA NY Harbor Health Care System, New York, New York, USA
| | - Herbert Lepor
- Department of Urology, New York University School of Medicine, New York, New York, USA
| | - James Wysock
- Department of Urology, New York University School of Medicine, New York, New York, USA
| | - Samir S Taneja
- Department of Urology, New York University School of Medicine, New York, New York, USA
| | - William C Huang
- Department of Urology, New York University School of Medicine, New York, New York, USA
| | - Daniel J Becker
- Department of Medicine, Laura & Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
- Department of Medicine, Manhattan Campus, VA NY Harbor Health Care System, New York, New York, USA
| | - Arjun V Balar
- Department of Medicine, Laura & Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Jonathan Melamed
- Department of Pathology, New York University School of Medicine, New York, New York, USA
| | - Fang-Ming Deng
- Department of Pathology, New York University School of Medicine, New York, New York, USA
| | - Qinghu Ren
- Department of Pathology, New York University School of Medicine, New York, New York, USA
| | - Donald Kufe
- Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Kwok-Kin Wong
- Department of Medicine, Laura & Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Dennis O Adeegbe
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Jiehui Deng
- Department of Medicine, Laura & Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - David R Wise
- Department of Medicine, Laura & Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| |
Collapse
|
2
|
Ouellet V, Erickson A, Wiley K, Morrissey C, Berge V, Moreno CS, Tasken KA, Trudel D, True LD, Lewis MS, Svindland A, Ertunc O, Vidal ID, Osunkoya AO, Jones T, Bova GS, Lamminen T, Achtman AH, Buzza M, Kouspou MM, Bigler SA, Zhou X, Freedland SJ, Mes-Masson AM, Garraway IP, Trock BJ, Taimen P, Saad F, Mirtti T, Knudsen BS, De Marzo AM. The Movember Global Action Plan 1 (GAP1): Unique Prostate Cancer Tissue Microarray Resource. Cancer Epidemiol Biomarkers Prev 2022; 31:715-727. [PMID: 35131885 PMCID: PMC9381093 DOI: 10.1158/1055-9965.epi-21-0600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/26/2021] [Accepted: 01/31/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The need to better understand the molecular underpinnings of the heterogeneous outcomes of patients with prostate cancer is a pressing global problem and a key research priority for Movember. To address this, the Movember Global Action Plan 1 Unique tissue microarray (GAP1-UTMA) project constructed a set of unique and richly annotated tissue microarrays (TMA) from prostate cancer samples obtained from multiple institutions across several global locations. METHODS Three separate TMA sets were built that differ by purpose and disease state. RESULTS The intended use of TMA1 (Primary Matched LN) is to validate biomarkers that help determine which clinically localized prostate cancers with associated lymph node metastasis have a high risk of progression to lethal castration-resistant metastatic disease, and to compare molecular properties of high-risk index lesions within the prostate to regional lymph node metastases resected at the time of prostatectomy. TMA2 (Pre vs. Post ADT) was designed to address questions regarding risk of castration-resistant prostate cancer (CRPC) and response to suppression of the androgen receptor/androgen axis, and characterization of the castration-resistant phenotype. TMA3 (CRPC Met Heterogeneity)'s intended use is to assess the heterogeneity of molecular markers across different anatomic sites in lethal prostate cancer metastases. CONCLUSIONS The GAP1-UTMA project has succeeded in combining a large set of tissue specimens from 501 patients with prostate cancer with rich clinical annotation. IMPACT This resource is now available to the prostate cancer community as a tool for biomarker validation to address important unanswered clinical questions around disease progression and response to treatment.
Collapse
Affiliation(s)
- Véronique Ouellet
- Centre de recherche du Centre hospitalier de l'Université de Montréal et Institut du cancer de Montréal, Montreal, Canada
| | - Andrew Erickson
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- Department of Pathology, Helsinki and Uusimaa Hospital District and Medicum, University of Helsinki, Helsinki, Finland
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Kathy Wiley
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, Washington
| | - Viktor Berge
- Department of Urology, Oslo University Hospital, Oslo, Norway
| | - Carlos S. Moreno
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Kristin Austlid Tasken
- Institute of Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Dominique Trudel
- Centre de recherche du Centre hospitalier de l'Université de Montréal et Institut du cancer de Montréal, Montreal, Canada
- Department of Pathology and Cellular Biology, Université de Montréal, Montreal, Canada
| | - Lawrence D. True
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Michael S. Lewis
- West Los Angeles Veterans Affairs Medical Center and Departments of Pathology and Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Aud Svindland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Onur Ertunc
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Suleyman Demirel University, Department of Pathology, Training and Research Hospital East Campus, Isparta, Turkey
| | - Igor Damasceno Vidal
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Adeboye O. Osunkoya
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Tracy Jones
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - G. Steven Bova
- Faculty of Medicine and Health Technology, Prostate Cancer Research Center, Tampere University and Tays Cancer Center, Tampere, Finland
| | - Tarja Lamminen
- Institute of Biomedicine, University of Turku, Turku, Finland
| | | | | | | | - Steven A. Bigler
- Department of Pathology, Mississippi Baptist Medical Center, Jackson, Mississippi
| | - Xinchun Zhou
- Department of Pathology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Stephen J. Freedland
- Center for Integrated Research on Cancer and Lifestyle, Cedars-Sinai Medical Center, Los Angeles, California
- Section of Urology, Durham VA Medical Center, Durham, North Carolina
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal et Institut du cancer de Montréal, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Isla P. Garraway
- Department of Urology, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California, Los Angeles, California
- Division of Urology, Greater Los Angeles VA Healthcare System, Los Angeles, California
| | - Bruce J. Trock
- Department of Urology and Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Pekka Taimen
- Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Pathology, Turku University Hospital, Turku, Finland
| | - Fred Saad
- Centre de recherche du Centre hospitalier de l'Université de Montréal et Institut du cancer de Montréal, Montreal, Canada
- Department of Surgery, Université de Montréal, Montreal, Canada
| | - Tuomas Mirtti
- HUS Diagnostic Center, Department of Pathology, HUS Helsinki University Hospital, Helsinki, Finland
- Medicum and Research Program In Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Beatrice S. Knudsen
- Digital and Computational Pathology, University of Utah, Salt Lake City, Utah
| | - Angelo M. De Marzo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Urology and Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| |
Collapse
|
3
|
Hempel Sullivan H, Maynard JP, Heaphy CM, Lu J, De Marzo AM, Lotan TL, Joshu CE, Sfanos KS. Differential mast cell phenotypes in benign versus cancer tissues and prostate cancer oncologic outcomes. J Pathol 2021; 253:415-426. [PMID: 33338262 DOI: 10.1002/path.5606] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 12/03/2020] [Accepted: 12/15/2020] [Indexed: 12/22/2022]
Abstract
We reported previously that high numbers of mast cells in benign (extra-tumoral) regions of the prostate are associated with worse outcomes after radical prostatectomy including biochemical recurrence and the development of metastases. Herein, with a cohort of 384 men, we performed mast cell subtyping and report that higher minimum number of the tryptase-only (MCT ) subset of extra-tumoral mast cells is associated with increased risk of biochemical recurrence (comparing highest to lowest tertiles: HR 2.32, 95% CI 1.37-3.93; P-trend = 0.002), metastases (HR 3.62, 95% CI 1.75-7.47; P-trend 0.001), and death from prostate cancer (HR 2.87, 95% CI 1.19-6.95; P-trend = 0.02). Preliminary RNA sequencing and comparison of benign versus cancer tissue mast cells revealed differential expression of additional site-specific genes. We further demonstrate that the genes CXCR4 and TFE3 are more highly expressed in tumor-infiltrating mast cells as well as other tumor-infiltrating immune cells and in tumor cells, respectively, and represent an altered tumor microenvironment. KIT variants were also differentially expressed in benign versus cancer tissue mast cells, with KIT variant 1 (GNNK+ ) mast cells identified as more prevalent in extra-tumoral regions of the prostate. Finally, using an established mouse model, we found that mast cells do not infiltrate Hi-Myc tumors, providing a model to specifically examine the role of extra-tumoral mast cells in tumorigenesis. Hi-Myc mice crossed to mast cell knockout (Wsh) mice and aged to 1 year revealed a higher degree of pre-invasive lesions and invasive cancer in wild-type mice versus heterozygous and knockout mice. This suggests a dosage effect where higher numbers of extra-tumoral mast cells resulted in higher cancer invasion. Overall, our studies provide further evidence for a role of extra-tumoral mast cells in driving adverse prostate cancer outcomes. © 2020 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Heidi Hempel Sullivan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Janielle P Maynard
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher M Heaphy
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Jiayun Lu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Angelo M De Marzo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.,Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Corinne E Joshu
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.,Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Karen S Sfanos
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.,Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
4
|
Wise DR, Schneider JA, Armenia J, Febles VA, McLaughlin B, Brennan R, Thoren KL, Abida W, Sfanos KS, De Marzo AM, Yegnasubramanian S, Fox JJ, Haas M, Heath H, Kagey MH, Newman W, Sirard CA, Fleisher M, Morris MJ, Chen Y, Larson SM, Haffner MC, Nelson PS, Schultz N, Garabedian MJ, Scher HI, Logan SK, Sawyers CL. Dickkopf-1 Can Lead to Immune Evasion in Metastatic Castration-Resistant Prostate Cancer. JCO Precis Oncol 2020; 4:2000097. [PMID: 33015525 DOI: 10.1200/po.20.00097] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2020] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Metastatic castration-resistant prostate cancer (mCRPC) with low androgen receptor (AR) and without neuroendocrine signaling, termed double-negative prostate cancer (DNPC), is increasingly prevalent in patients treated with AR signaling inhibitors and is in need of new biomarkers and therapeutic targets. METHODS Candidate genes enriched in DNPC were determined using differential gene expression analysis of discovery and validation cohorts of mCRPC biopsies. Laboratory studies were carried out in human mCRPC organoid cultures, prostate cancer (PCa) cell lines, and mouse xenograft models. Epigenetic studies were carried out in a rapid autopsy cohort. RESULTS Dickkopf-1 (DKK1) expression is increased in DNPC relative to prostate-specific antigen (PSA)-expressing mCRPC in the Stand Up to Cancer/Prostate Cancer Foundation discovery cohort (11.2 v 0.28 reads per kilobase per million mapped reads; q < 0.05; n = 117) and in the University of Washington/Fred Hutchinson Cancer Research Center cohort (9.2 v 0.99 fragments per kilobase of transcript per million mapped reads; P < .0001). DKK1 expression can be regulated by activated Wnt signaling in vitro and correlates with activating canonical Wnt signaling mutations and low PSA mRNA in mCRPC biopsies (P < .05). DKK1 hypomethylation was associated with increased DKK1 mRNA expression (Pearson r = -0.66; P < .0001) in a rapid autopsy cohort (n = 7). DKK1-high mCRPC biopsies are infiltrated with significantly higher numbers of quiescent natural killer (NK) cells (P < .005) and lower numbers of activated NK cells (P < .0005). Growth inhibition of the human PCa model PC3 by the anti-DKK1 monoclonal antibody DKN-01 depends on the presence of NK cells in a severe combined immunodeficient xenograft mouse model. CONCLUSION These results support DKK1 as a contributor to the immunosuppressive tumor microenvironment of DNPC. These data have provided the rationale for a clinical trial targeting DKK1 in mCRPC (ClinicalTrials.gov identifier: NCT03837353).
Collapse
Affiliation(s)
- David R Wise
- Department of Medicine, Perlmutter Cancer Center, NYU Langone Medical Center, New York, NY
| | | | - Joshua Armenia
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Victor Adorno Febles
- Department of Medicine, Perlmutter Cancer Center, NYU Langone Medical Center, New York, NY
| | - Bridget McLaughlin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ryan Brennan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Katie L Thoren
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Wassim Abida
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Karen S Sfanos
- Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD.,Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD.,Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD
| | - Angelo M De Marzo
- Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD.,Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD.,Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD
| | - Srinivasan Yegnasubramanian
- Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD.,Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD
| | - Josef J Fox
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | | | | | | - Martin Fleisher
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael J Morris
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yu Chen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Steven M Larson
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael C Haffner
- Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD.,Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD
| | - Peter S Nelson
- Departments of Medicine and Pathology, University of Washington, and Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Nikolaus Schultz
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael J Garabedian
- Department of Urology, NYU Langone Medical Center, New York, NY.,Department of Microbiology, NYU Langone Medical Center, New York, NY
| | - Howard I Scher
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Susan K Logan
- Department of Urology, NYU Langone Medical Center, New York, NY
| | - Charles L Sawyers
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY.,Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | | |
Collapse
|
5
|
Fast prostate retrieval in robot-assisted laparoscopic prostatectomy for next-generation biobanking. J Robot Surg 2019; 14:271-274. [PMID: 31129776 DOI: 10.1007/s11701-019-00974-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 05/14/2019] [Indexed: 10/26/2022]
Abstract
Robotic-assisted laparoscopic radical prostatectomy (RALP) has become the most widespread treatment for organ-confined prostate cancer. Here, we describe a fast specimen retrieval technique for RALP to obtain high-quality tissue specimen with minimal warm ischemia time for next-generation biobanking. Here, we show that using fast retrieval technique, short warm ischemia times can be achieved while not increasing the surgical time. Patients undergoing RALP with written informed consent participated in Helsinki Urological Bank study. Previously operated RALP patients and those, who were not willing to participate in the study, served as a control group. The study consisted of 1685 patients, 684 in fast retrieval and 1001 in control group. We developed a novel fast retrieval technique in which fascia is opened for camera port according to the prostate size and a running suture is placed and tightened against the camera port in the beginning of the operation. Immediately after prostate is freed from attachments, suture is loosened and the prostate is extirpated inside the endoscopic bag through the camera port fascial opening, then the fascial suture is again tightened against the camera port and the RALP procedure is completed. The mean warm ischemia times in fast retrieval group were 20 min 18 s and 22 min 30 s, respectively, in patients without and with lymphadenectomy. The mean console and surgery times with and without lymphadenectomy were similar in both groups. There were no technique-related complications associated with Fast Retrieval procedure. Tissue integrity test results for the RNA and DNA quality showed good quality for the specimen. Fast retrieval technique can easily and safely be utilized to maximize usefulness of RALP tissue specimen in downstream biobank applications.
Collapse
|
6
|
Hulsen T. An overview of publicly available patient-centered prostate cancer datasets. Transl Androl Urol 2019; 8:S64-S77. [PMID: 31143673 PMCID: PMC6511704 DOI: 10.21037/tau.2019.03.01] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/27/2019] [Indexed: 02/05/2023] Open
Abstract
Prostate cancer (PCa) is the second most common cancer in men, and the second leading cause of death from cancer in men. Many studies on PCa have been carried out, each taking much time before the data is collected and ready to be analyzed. However, on the internet there is already a wide range of PCa datasets available, which could be used for data mining, predictive modelling or other purposes, reducing the need to setup new studies to collect data. In the current scientific climate, moving more and more to the analysis of "big data" and large, international, multi-site projects using a modern IT infrastructure, these datasets could be proven extremely valuable. This review presents an overview of publicly available patient-centered PCa datasets, divided into three categories (clinical, genomics and imaging) and an "overall" section to enable researchers to select a suitable dataset for analysis, without having to go through days of work to find the right data. To acquire a list of human PCa databases, scientific literature databases and academic social network sites were searched. We also used the information from other reviews. All databases in the combined list were then checked for public availability. Only databases that were either directly publicly available or available after signing a research data agreement or retrieving a free login were selected for inclusion in this review. Data should be available to commercial parties as well. This paper focuses on patient-centered data, so the genomics data section does not include gene-centered databases or pathway-centered databases. We identified 42 publicly available, patient-centered PCa datasets. Some of these consist of different smaller datasets. Some of them contain combinations of datasets from the three data domains: clinical data, imaging data and genomics data. Only one dataset contains information from all three domains. This review presents all datasets and their characteristics: number of subjects, clinical fields, imaging modalities, expression data, mutation data, biomarker measurements, etc. Despite all the attention that has been given to making this overview of publicly available databases as extensive as possible, it is very likely not complete, and will also be outdated soon. However, this review might help many PCa researchers to find suitable datasets to answer the research question with, without the need to start a new data collection project. In the coming era of big data analysis, overviews like this are becoming more and more useful.
Collapse
Affiliation(s)
- Tim Hulsen
- Department of Professional Health Solutions & Services, Philips Research, Eindhoven, The Netherlands
| |
Collapse
|
7
|
Baena-Del Valle JA, Zheng Q, Esopi DM, Rubenstein M, Hubbard GK, Moncaliano MC, Hruszkewycz A, Vaghasia A, Yegnasubramanian S, Wheelan SJ, Meeker AK, Heaphy CM, Graham MK, De Marzo AM. MYC drives overexpression of telomerase RNA (hTR/TERC) in prostate cancer. J Pathol 2017; 244:11-24. [PMID: 28888037 DOI: 10.1002/path.4980] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 08/07/2017] [Accepted: 08/24/2017] [Indexed: 01/21/2023]
Abstract
Telomerase consists of at least two essential elements, an RNA component hTR or TERC that contains the template for telomere DNA addition and a catalytic reverse transcriptase (TERT). While expression of TERT has been considered the key rate-limiting component for telomerase activity, increasing evidence suggests an important role for the regulation of TERC in telomere maintenance and perhaps other functions in human cancer. By using three orthogonal methods including RNAseq, RT-qPCR, and an analytically validated chromogenic RNA in situ hybridization assay, we report consistent overexpression of TERC in prostate cancer. This overexpression occurs at the precursor stage (e.g. high-grade prostatic intraepithelial neoplasia or PIN) and persists throughout all stages of disease progression. Levels of TERC correlate with levels of MYC (a known driver of prostate cancer) in clinical samples and we also show the following: forced reductions of MYC result in decreased TERC levels in eight cancer cell lines (prostate, lung, breast, and colorectal); forced overexpression of MYC in PCa cell lines, and in the mouse prostate, results in increased TERC levels; human TERC promoter activity is decreased after MYC silencing; and MYC occupies the TERC locus as assessed by chromatin immunoprecipitation (ChIP). Finally, we show that knockdown of TERC by siRNA results in reduced proliferation of prostate cancer cell lines. These studies indicate that TERC is consistently overexpressed in all stages of prostatic adenocarcinoma and that its expression is regulated by MYC. These findings nominate TERC as a novel prostate cancer biomarker and therapeutic target. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Javier A Baena-Del Valle
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Pathology and Laboratory Medicine, Fundacion Santa Fe De Bogota University Hospital, Bogota, DC, Colombia
| | - Qizhi Zheng
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David M Esopi
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael Rubenstein
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, Maryland, USA
| | - Gretchen K Hubbard
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Maria C Moncaliano
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Hruszkewycz
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland, USA
| | - Ajay Vaghasia
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Srinivasan Yegnasubramanian
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Departments of Urology and Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Brady Urological Research Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sarah J Wheelan
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Departments of Urology and Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Brady Urological Research Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alan K Meeker
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Brady Urological Research Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Christopher M Heaphy
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Brady Urological Research Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mindy K Graham
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Brady Urological Research Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Angelo M De Marzo
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Departments of Urology and Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Brady Urological Research Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Loss of miR-26a-5p promotes proliferation, migration, and invasion in prostate cancer through negatively regulating SERBP1. Tumour Biol 2016; 37:12843-12854. [PMID: 27449037 DOI: 10.1007/s13277-016-5158-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 07/12/2016] [Indexed: 01/07/2023] Open
Abstract
The biological role of miR-26a involved in the carcinogenesis of prostate cancer (PC) has been controversial. Besides, the underlying mechanism by which miR-26a plays a role in PC has been unclear. To investigate the role of miR-26a-5p in the PC, miR-26a-5p was detected and statistically analyzed in clinical PC tissues and a panel of PC cell lines. Using bioinformatics analysis, we found that serpine1 messenger RNA (mRNA) binding protein 1 (SERBP1) was a potential downstream target of miR-26a-5p. Using luciferase reporter and western blot, we identified that miR-26a-5p negatively regulated SERBP1 on the PC cell line level. It was confirmed that miR-26a-5p was markedly downregulated in PC tissues compared with normal controls whose reduced expression was significantly associated with metastasis and poor overall prognosis and found that miR-26a-5p was able to prevent proliferation and motility of PC cells in vitro. Additionally, SERBP1 was identified as a downstream target of miR-26a-5p. Moreover, it was observed that SERBP1 was markedly upregulated in prostate cancer tissues and was significantly associated with tissue metastasis and Gleason score. Taken together, our results for the first time demonstrate that the loss of miR-26a-5p promotes proliferation, migration, and invasion through targeting SERBP1 in PC, supporting the tumor-suppressing role of miR-26a-5p in PC.
Collapse
|
9
|
Yu SH, Zheng Q, Esopi D, Macgregor-Das A, Luo J, Antonarakis ES, Drake CG, Vessella R, Morrissey C, De Marzo AM, Sfanos KS. A Paracrine Role for IL6 in Prostate Cancer Patients: Lack of Production by Primary or Metastatic Tumor Cells. Cancer Immunol Res 2015; 3:1175-84. [PMID: 26048576 DOI: 10.1158/2326-6066.cir-15-0013] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 05/26/2015] [Indexed: 01/01/2023]
Abstract
Correlative human studies suggest that the pleiotropic cytokine IL6 contributes to the development and/or progression of prostate cancer. However, the source of IL6 production in the prostate microenvironment in patients has yet to be determined. The cellular origin of IL6 in primary and metastatic prostate cancer was examined in formalin-fixed, paraffin-embedded tissues using a highly sensitive and specific chromogenic in situ hybridization (CISH) assay that underwent extensive analytical validation. Quantitative RT-PCR showed that benign prostate tissues often had higher expression of IL6 mRNA than matched tumor specimens. CISH analysis further indicated that both primary and metastatic prostate adenocarcinoma cells do not express IL6 mRNA. IL6 expression was highly heterogeneous across specimens and was nearly exclusively restricted to the prostate stromal compartment--including endothelial cells and macrophages, among other cell types. The number of IL6-expressing cells correlated positively with the presence of acute inflammation. In metastatic disease, tumor cells were negative in all lesions examined, and IL6 expression was restricted to endothelial cells within the vasculature of bone metastases. Finally, IL6 was not detected in any cells in soft tissue metastases. These data suggest that, in prostate cancer patients, paracrine rather than autocrine IL6 production is likely associated with any role for the cytokine in disease progression.
Collapse
Affiliation(s)
- Shu-Han Yu
- Department of Pathology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Qizhi Zheng
- Department of Pathology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - David Esopi
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anne Macgregor-Das
- Department of Pathology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jun Luo
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Emmanuel S Antonarakis
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Charles G Drake
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert Vessella
- Department of Urology, University of Washington Medical Center, Seattle, Washington
| | - Colm Morrissey
- Department of Urology, University of Washington Medical Center, Seattle, Washington
| | - Angelo M De Marzo
- Department of Pathology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland. Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Karen S Sfanos
- Department of Pathology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland. Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
10
|
Liang J, Li Y, Daniels G, Sfanos K, De Marzo A, Wei J, Li X, Chen W, Wang J, Zhong X, Melamed J, Zhao J, Lee P. LEF1 Targeting EMT in Prostate Cancer Invasion Is Regulated by miR-34a. Mol Cancer Res 2015; 13:681-8. [PMID: 25587085 DOI: 10.1158/1541-7786.mcr-14-0503] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 12/30/2014] [Indexed: 12/16/2022]
Abstract
UNLABELLED The microRNA-34a (miR-34a), a tumor-suppressive microRNA (miRNA), is implicated in epithelial-mesenchymal transition (EMT) and cancer stem cells. Lymphoid enhancer-binding factor-1 (LEF1) is a key transcription factor in the Wnt signaling pathway, and has been suggested to be involved in regulation of cell proliferation and invasion. Here, the molecular mechanism of miR-34a and LEF1 in cooperatively regulating prostate cancer cell invasion is described. Molecular profiling analysis of miRNA levels in prostate cancer cells revealed a negative correlation between miR-34a and LEF1 expression, and the downregulation of LEF1 by miR-34a was confirmed by luciferase assays. Furthermore, miR-34a specifically repressed LEF1 expression through direct binding to its 3'-untranslated regions (3'-UTR). miR-34a modulated the levels of LEF1 to regulate EMT in prostate cancer cells. Functionally, miR-34a negatively correlated with the migration and invasion of prostate cancer cells through LEF1. An analysis of miR-34a expression levels in matched human tumor and benign tissues demonstrated consistent and statistically significant downregulation of miR-34a in primary prostate cancer specimens. These data strongly suggest that miR-34a/LEF1 regulation of EMT plays an important role in prostate cancer migration and invasion. IMPLICATIONS The miR-34a-LEF1 axis represents a potential molecular target for novel therapeutic strategies in prostate cancer.
Collapse
Affiliation(s)
- Jiaqian Liang
- Department of Pathology, New York University School of Medicine, New York, New York. Department of Urology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yirong Li
- Department of Pathology, New York University School of Medicine, New York, New York
| | - Garrett Daniels
- Department of Pathology, New York University School of Medicine, New York, New York
| | - Karen Sfanos
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Angelo De Marzo
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Jianjun Wei
- Department of Pathology, Northwestern University, Chicago, Illinois
| | - Xin Li
- NYU Cancer Institute, New York University School of Medicine, New York, New York. Department of Urology, New York University School of Medicine, New York, New York. Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York
| | - Wenqiang Chen
- Pediatric Lab of Medical Science Experiment Center, Guangxi Medical University, Nanning, Guangxi, China
| | - Jinhua Wang
- NYU Cancer Institute, New York University School of Medicine, New York, New York
| | - Xuelin Zhong
- Department of Pathology, New York University School of Medicine, New York, New York
| | - Jonathan Melamed
- Department of Pathology, New York University School of Medicine, New York, New York
| | - Jun Zhao
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Peng Lee
- Department of Pathology, New York University School of Medicine, New York, New York. NYU Cancer Institute, New York University School of Medicine, New York, New York. Department of Urology, New York University School of Medicine, New York, New York. New York Harbor Healthcare System, New York University School of Medicine, New York, New York.
| |
Collapse
|
11
|
Zheng Q, Peskoe SB, Ribas J, Rafiqi F, Kudrolli T, Meeker AK, De Marzo AM, Platz EA, Lupold SE. Investigation of miR-21, miR-141, and miR-221 expression levels in prostate adenocarcinoma for associated risk of recurrence after radical prostatectomy. Prostate 2014; 74:1655-62. [PMID: 25252191 PMCID: PMC4205269 DOI: 10.1002/pros.22883] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 07/28/2014] [Indexed: 12/29/2022]
Abstract
BACKGROUND MicroRNAs (miRNAs) are small non-coding RNAs that regulate a broad array of cellular and disease processes. Several miRNAs are differentially expressed in cancer and many are being considered as biomarkers for predicting clinical outcomes. Here we quantified the expression of three miRNAs, miR-21, miR-141, and miR-221, from prostate cancer surgical specimens and evaluated their association with disease recurrence after primary therapy. METHODS A pilot nested case-control study was designed from a large cohort of men who underwent radical prostatectomy between 1993 and 2001. Total RNA was extracted from malignant prostate tissue of 59 cases (recurrence) and 59 controls. Cases and controls were matched on age, race, pathologic stage, and grade. The relative expression of each miRNA was then determined for each sample by quantitative real-time RT-PCR. Conditional logistic regression was used to estimate odds ratios (OR) and 95% confidence intervals (CI) of recurrence for tertiles of miRNA expression. We noted block storage time effects and thus, used separate tertile cutpoints based on the controls by calendar year of prostatectomy. RESULTS Lower miR-221 expression was associated with a higher risk of recurrence; the ORs were 3.21 for the lowest tertile and 2.63 for the middle tertile compared with the highest tertile of expression (P-trend = 0.02). This pattern was unchanged after multivariable adjustment (P-trend = 0.05). No statistically significant trends were observed for miR-21 or miR-141 after multivariable adjustment. CONCLUSIONS Based on this small pilot study, men with localized prostate cancers with lower miR-221 expression may have a greater risk for recurrence after surgery.
Collapse
Affiliation(s)
- Qizhi Zheng
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Sarah B. Peskoe
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Judit Ribas
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Fatema Rafiqi
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Tarana Kudrolli
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Alan K. Meeker
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Angelo M. De Marzo
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Elizabeth A. Platz
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Shawn E. Lupold
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
- Correspondence: ; 600 N Wolfe St, Park 209, Baltimore, MD 21287 Phone: 410-502-4822, FAX: 410-502-7711
| |
Collapse
|
12
|
Ragin C, Park JY. Biospecimens, biobanking and global cancer research collaborations. Ecancermedicalscience 2014; 8:454. [PMID: 25228910 PMCID: PMC4154946 DOI: 10.3332/ecancer.2014.454] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Indexed: 11/08/2022] Open
Abstract
The disparities in prostate cancer incidence and mortality continue to be a global public health problem. Efforts to address the prostate cancer disparity in black men have been met with a number of challenges, specifically in the accessibility to biospecimens in the context of global prostate cancer collaborations. During the International Educational Workshop at the Science of Global Prostate Cancer Disparities conference held 1–4 November 2012 in Nassau, the Bahamas, an overview of biobanking and biospecimen repositories, and materials transfer in global prostate cancer collaborations were discussed. The challenges faced by low-resource countries were identified, and potential solutions were recommended.
Collapse
Affiliation(s)
- Camille Ragin
- Cancer Prevention and Control Programme, Fox Chase Cancer Centre, Temple University Health System, 333 Cottman Avenue, Philadelphia, PA 19111, USA ; African Caribbean Cancer Consortium (AC3)
| | - Jong Y Park
- African Caribbean Cancer Consortium (AC3) ; Department of Cancer Epidemiology, Moffitt Cancer Centre, Tampa, FL 33612, USA ; Prostate Cancer Transatlantic Consortium (CaPTC)
| |
Collapse
|