1
|
Lin YY, Noghabi HS, Volik S, Bell R, Sar F, Haegert A, Chung HC, Fazli L, Oo HZ, Daugaard M, Kuo MH, Hsu SC, Imeda EL, Zanettini C, Queiroz L, Schlotmann B, Gheybi K, Cooper C, Kote-Jarai Z, Eeles R, Kung HJ, Marchionni L, Weischenfeldt J, Miller KD, Rabinowitz A, Wang Y, Zhang HF, Sorensen PH, Carey MS, Gleave M, Hayes VM, Gibson WT, Collins CC. Identifying Rare Germline Variants Associated with Metastatic Prostate Cancer Through an Extreme Phenotype Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.28.25326584. [PMID: 40343042 PMCID: PMC12060958 DOI: 10.1101/2025.04.28.25326584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Background Studies of germline variants in prostate cancer (PCa) have largely focused on their connections to cancer predisposition. However, an understanding of how heritable factors contribute to cancer progression and metastasis remain limited. Objective To identify low frequency to rare germline nonsynonymous variants associated with increased risk for metastatic PCa (mPCa), while providing functional validation. Design We assembled an extreme phenotype cohort (EPC) of 52 patients diagnosed with predominantly high-grade (Gleason Score (GS) ≥ 8) PCa and > 7 years of follow-up for which localized treatment naïve tumor tissues were available. In half of the cases, the tumor had metastasized to bone, providing an even distribution of bone mPCa and non mPCa cases. Tumor and matched distant benign DNA samples were exome sequenced and analyzed for germline variants with population-wide minor allelic frequencies ≤ 2%. Findings were validated using two independent PCa germline cohorts, including a closely matched Australian study biased to aggressive disease (n = 53) and Pan Prostate Cancer Group (PPCG, n = 976). Two mPCa-promoting candidate variants in KDM6B and BRCA2 were engineered into cell lines and functionalized. Results Germline nonsynonymous rare variants (gnsRVs) identified in 25 DNA Damage Repair (DDR) genes were significantly enriched in the mPCa patients (p=4.57e-06). Conversely, the prevalence of synonymous variants at minor allele frequencies of ≤ 2% were similar between the mPCa and non mPCa patients. The predictive power of variants in 53 non-DDR genes was validated in the Australian cohort (p=0.028) and correlated with high-risk PCa in PPCG (p=0.03). KDM6B K973Q showed functional significance despite being annotated as benign in ClinVar, while BRCA2 I1962T showed sensitivity to Olaparib. In total, six EPC variants related to DNA repair or epigenetics were found to alter enzymatic activity. Conclusions EPCs coupled with low frequency/rare variant analyses may advance understanding of interactions between the germline and tumor in PCa. We identified a series of germline variants that were enriched among mPCa patients. Moreover, we showed that one of these variants confers a metastatic phenotype. Our findings suggest that germline testing at diagnosis may improve treatment stratification in PCa. Patient summary The presence of specific genetic variants among men with PCa may elevate the risk of mPCa once PCa develops. Knowledge of the variant burden at time of diagnosis may enable accurate stratification of some patients for aggressive therapeutic interventions.
Collapse
Affiliation(s)
- Yen-Yi Lin
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
- These authors are joint first authors
| | - Hamideh Sharifi Noghabi
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
- These authors are joint first authors
| | - Stanislav Volik
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Robert Bell
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Funda Sar
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Anne Haegert
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Hee Chul Chung
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Ladan Fazli
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Htoo Zarni Oo
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Mads Daugaard
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Ming-Han Kuo
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Sheng-Chieh Hsu
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Eddie L Imeda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, USA
| | - Claudio Zanettini
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, USA
| | - Lucio Queiroz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, USA
| | - Balthasar Schlotmann
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
| | - Kazzem Gheybi
- Ancestry and Health Genomics Laboratory, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - Colin Cooper
- The Institute of Cancer Research, London, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Zsofia Kote-Jarai
- The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, London, UK
| | - Rosalind Eeles
- The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, London, UK
| | - Hsing-Jien Kung
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Luigi Marchionni
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, USA
| | - Joachim Weischenfeldt
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
- Department of Urology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Alan Rabinowitz
- Rural Coordination Center of British Columbia, Vancouver, British Columbia, Canada
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
- Department of Experimental Therapeutics, BC Cancer, Vancouver, British Columbia, Canada
| | - Hai-Feng Zhang
- Department of Molecular Oncology, BC Cancer, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Poul H Sorensen
- Department of Molecular Oncology, BC Cancer, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mark S Carey
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, Canada
| | - Martin Gleave
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Vanessa M Hayes
- Ancestry and Health Genomics Laboratory, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
- School of Health Systems and Public Health, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | - William T Gibson
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- These authors are joint last authors
| | - Colin C Collins
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
- These authors are joint last authors
| |
Collapse
|
2
|
Di Giovannantonio M, Hartley F, Elshenawy B, Barberis A, Hudson D, Shafique HS, Allott VES, Harris DA, Lord SR, Haider S, Harris AL, Buffa FM, Harris BHL. Defining hypoxia in cancer: A landmark evaluation of hypoxia gene expression signatures. CELL GENOMICS 2025; 5:100764. [PMID: 39892389 PMCID: PMC11872601 DOI: 10.1016/j.xgen.2025.100764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/04/2024] [Accepted: 01/07/2025] [Indexed: 02/03/2025]
Abstract
Tumor hypoxia drives metabolic shifts, cancer progression, and therapeutic resistance. Challenges in quantifying hypoxia have hindered the exploitation of this potential "Achilles' heel." While gene expression signatures have shown promise as surrogate measures of hypoxia, signature usage is heterogeneous and debated. Here, we present a systematic pan-cancer evaluation of 70 hypoxia signatures and 14 summary scores in 104 cell lines and 5,407 tumor samples using 472 million length-matched random gene signatures. Signature and score choice strongly influenced the prediction of hypoxia in vitro and in vivo. In cell lines, the Tardon signature was highly accurate in both bulk and single-cell data (94% accuracy, interquartile mean). In tumors, the Buffa and Ragnum signatures demonstrated superior performance, with Buffa/mean and Ragnum/interquartile mean emerging as the most promising for prospective clinical trials. This work delivers recommendations for experimental hypoxia detection and patient stratification for hypoxia-targeting therapies, alongside a generalizable framework for signature evaluation.
Collapse
Affiliation(s)
- Matteo Di Giovannantonio
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK
| | - Fiona Hartley
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK
| | - Badran Elshenawy
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK
| | - Alessandro Barberis
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK
| | - Dan Hudson
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK; The Rosalind Franklin Institute, Didcot, UK
| | | | | | | | - Simon R Lord
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK
| | - Syed Haider
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - Adrian L Harris
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK
| | - Francesca M Buffa
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK; CompBio Lab, Department of Computing Sciences, Bocconi University, Milan, Italy; AI and Systems Biology Lab, IFOM - Istituto Fondazione di Oncologia Molecolare ETS, Milan, Italy.
| | - Benjamin H L Harris
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK; St. Catherine's College, University of Oxford, Oxford, UK; Cutrale Perioperative and Ageing Group, Imperial College London, London, UK.
| |
Collapse
|
3
|
Pakula H, Pederzoli F, Fanelli GN, Nuzzo PV, Rodrigues S, Loda M. Deciphering the Tumor Microenvironment in Prostate Cancer: A Focus on the Stromal Component. Cancers (Basel) 2024; 16:3685. [PMID: 39518123 PMCID: PMC11544791 DOI: 10.3390/cancers16213685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Prostate cancer progression is significantly affected by its tumor microenvironment, in which mesenchymal cells play a crucial role. Stromal cells are modified by cancer mutations, response to androgens, and lineage plasticity, and in turn, engage with epithelial tumor cells via a complex array of signaling pathways and ligand-receptor interactions, ultimately affecting tumor growth, immune interaction, and response to therapy. The metabolic rewiring and interplay in the microenvironment play an additional role in affecting the growth and progression of prostate cancer. Finally, therapeutic strategies and novel clinical trials with agents that target the stromal microenvironment or disrupt the interaction between cellular compartments are described. This review underscores cancer-associated fibroblasts as essential contributors to prostate cancer biology, emphasizing their potential as prognostic indicators and therapeutic targets.
Collapse
Affiliation(s)
- Hubert Pakula
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA; (H.P.); (F.P.); (G.N.F.); (P.V.N.); (S.R.)
| | - Filippo Pederzoli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA; (H.P.); (F.P.); (G.N.F.); (P.V.N.); (S.R.)
| | - Giuseppe Nicolò Fanelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA; (H.P.); (F.P.); (G.N.F.); (P.V.N.); (S.R.)
| | - Pier Vitale Nuzzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA; (H.P.); (F.P.); (G.N.F.); (P.V.N.); (S.R.)
| | - Silvia Rodrigues
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA; (H.P.); (F.P.); (G.N.F.); (P.V.N.); (S.R.)
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA; (H.P.); (F.P.); (G.N.F.); (P.V.N.); (S.R.)
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, New York, NY 10021, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave, Boston, MA 02215, USA
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX1 2JD, UK
| |
Collapse
|
4
|
Ziemann M, Schroeter B, Bora A. Two subtle problems with overrepresentation analysis. BIOINFORMATICS ADVANCES 2024; 4:vbae159. [PMID: 39539946 PMCID: PMC11557902 DOI: 10.1093/bioadv/vbae159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/21/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Motivation Overrepresentation analysis (ORA) is used widely to assess the enrichment of functional categories in a gene list compared to a background list. ORA is therefore a critical method in the interpretation of 'omics data, relating gene lists to biological functions and themes. Although ORA is hugely popular, we and others have noticed two potentially undesired behaviours of some ORA tools. The first one we call the 'background problem', because it involves the software eliminating large numbers of genes from the background list if they are not annotated as belonging to any category. The second one we call the 'false discovery rate problem', because some tools underestimate the true number of parallel tests conducted. Results Here, we demonstrate the impact of these issues on several real RNA-seq datasets and use simulated RNA-seq data to quantify the impact of these problems. We show that the severity of these problems depends on the gene set library, the number of genes in the list, and the degree of noise in the dataset. These problems can be mitigated by changing packages/websites for ORA or by changing to another approach such as functional class scoring. Availability and implementation An R/Shiny tool has been provided at https://oratool.ziemann-lab.net/ and the supporting materials are available from Zenodo (https://zenodo.org/records/13823301).
Collapse
Affiliation(s)
- Mark Ziemann
- Bioinformatics Working Group, Burnet Institute, Melbourne, VIC 3004, Australia
- School of Life and Environmental Sciences, Deakin University, Geelong, VIC 3216, Australia
| | - Barry Schroeter
- School of Life and Environmental Sciences, Deakin University, Geelong, VIC 3216, Australia
| | - Anusuiya Bora
- Bioinformatics Working Group, Burnet Institute, Melbourne, VIC 3004, Australia
- School of Life and Environmental Sciences, Deakin University, Geelong, VIC 3216, Australia
| |
Collapse
|
5
|
Heimdörfer D, Artamonova N, Culig Z, Heidegger I. Unraveling molecular characteristics and tumor microenvironment dynamics of neuroendocrine prostate cancer. J Cancer Res Clin Oncol 2024; 150:462. [PMID: 39412660 PMCID: PMC11485041 DOI: 10.1007/s00432-024-05983-0] [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: 06/30/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024]
Abstract
Prostate cancer (PCa) is the most prevalent malignancy and the second leading cause of cancer-related deaths among men. While adenocarcinoma of the prostate (adeno-PCa) is well-characterized, neuroendocrine prostate cancer (NEPC) remains poorly understood. Generally, NEPC is a rare but highly aggressive histological variant, however its limited patho-physiological understanding leads to insufficient treatment options associated with low survival rates for NEPC patients. Current treatments for NEPC, including platinum-based therapies, offer some efficacy, but there is a significant need for more targeted approaches. This review summarizes the molecular characteristics of NEPC in contrast to adeno-PCa, providing a comprehensive comparison. A significant portion of the discussion is dedicated to the tumor microenvironment (TME), which has recently been identified as a key factor in tumor progression. The TME includes various cells, signaling molecules, and the extracellular matrix surrounding the tumor, all of which play critical roles in cancer development and response to treatment. Understanding the TME's influence on NEPC could uncover new avenues for innovative treatment strategies, potentially improving outcomes for patients with this challenging variant of PCa.
Collapse
Affiliation(s)
- David Heimdörfer
- Department of Urology, Medical University Innsbruck, Innsbruck, Anichstreet 35, Innsbruck, A-6020, Austria
| | - Nastasiia Artamonova
- Department of Urology, Medical University Innsbruck, Innsbruck, Anichstreet 35, Innsbruck, A-6020, Austria
| | - Zoran Culig
- Department of Urology, Medical University Innsbruck, Innsbruck, Anichstreet 35, Innsbruck, A-6020, Austria
| | - Isabel Heidegger
- Department of Urology, Medical University Innsbruck, Innsbruck, Anichstreet 35, Innsbruck, A-6020, Austria.
| |
Collapse
|
6
|
Vecchiotti D, Clementi L, Cornacchia E, Di Vito Nolfi M, Verzella D, Capece D, Zazzeroni F, Angelucci A. Evidence of the Link between Stroma Remodeling and Prostate Cancer Prognosis. Cancers (Basel) 2024; 16:3215. [PMID: 39335188 PMCID: PMC11430343 DOI: 10.3390/cancers16183215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Prostate cancer (PCa), the most commonly diagnosed cancer in men worldwide, is particularly challenging for oncologists when a precise prognosis needs to be established. Indeed, the entire clinical management in PCa has important drawbacks, generating an intense debate concerning the possibility to individuate molecular biomarkers able to avoid overtreatment in patients with pathological indolent cancers. To date, the paradigmatic change in the view of cancer pathogenesis prompts to look for prognostic biomarkers not only in cancer epithelial cells but also in the tumor microenvironment. PCa ecology has been defined with increasing details in the last few years, and a number of promising key markers associated with the reactive stroma are now available. Here, we provide an updated description of the most biologically significant and cited prognosis-oriented microenvironment biomarkers derived from the main reactive processes during PCa pathogenesis: tissue adaptations, inflammatory response and metabolic reprogramming. Proposed biomarkers include factors involved in stromal cell differentiation, cancer-normal cell crosstalk, angiogenesis, extracellular matrix remodeling and energy metabolism.
Collapse
Affiliation(s)
- Davide Vecchiotti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Letizia Clementi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Emanuele Cornacchia
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Mauro Di Vito Nolfi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Daniela Verzella
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Daria Capece
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Francesca Zazzeroni
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Adriano Angelucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| |
Collapse
|
7
|
Wu HL, Wang XB, Li J, Zheng BW. The tumor-stroma ratio in giant cell tumor of bone: associations with the immune microenvironment and responsiveness to denosumab treatment. J Orthop Surg Res 2024; 19:405. [PMID: 39010095 PMCID: PMC11250954 DOI: 10.1186/s13018-024-04885-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 06/27/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Currently, there is limited understanding regarding the clinical significance of the tumor-stroma ratio (TSR) in giant cell tumor of bone (GCTB). Hence, we aimed to investigate the distribution of TSR in GCTB and explore its correlation with various clinicopathologic factors, immune microenvironment, survival prognosis, and denosumab treatment responsiveness. METHODS We conducted a multicenter cohort study comprising 426 GCTB patients treated at four centers. TSR was evaluated on hematoxylin and eosin-stained and immunofluorescent sections of tumor specimens. Immunohistochemistry was performed to assess CD3+, CD4+, CD8+, CD20+, PD-1+, PD-L1+, and FoxP3+ TIL subtypes as well as Ki-67 expression levels in 426 tissue specimens. These parameters were then analyzed for their correlations with patient outcomes [local recurrence-free survival (LRFS) and overall survival (OS)], clinicopathological features, and denosumab treatment responsiveness. RESULTS Low TSR was significantly associated with poor LRFS and OS in both cohorts. Furthermore, TSR was also correlated with multiple clinicopathological features, TIL subtype expression, and denosumab treatment responsiveness. TSR demonstrated similar predictive capabilities as the conventional Campanacci staging system for predicting patients' LRFS and OS. CONCLUSION The results of this study provide evidence supporting the use of TSR as a reliable prognostic tool in GCTB and as a predictor of denosumab treatment responsiveness. These findings may aid in developing individualized treatment strategies for GCTB patients in the future.
Collapse
Affiliation(s)
- Hai-Lin Wu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China
| | - Xiao-Bin Wang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China
| | - Jing Li
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China.
| | - Bo-Wen Zheng
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China.
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, No. 11, Xizhimen South Street, Xicheng District, Beijing, 100044, China.
| |
Collapse
|
8
|
Kader A, Snellings J, Adams LC, Gottheil P, Mangarova DB, Heyl JL, Kaufmann JO, Moeckel J, Brangsch J, Auer TA, Collettini F, Sauer F, Hamm B, Käs J, Sack I, Makowski MR, Braun J. Sensitivity of magnetic resonance elastography to extracellular matrix and cell motility in human prostate cancer cell line-derived xenograft models. BIOMATERIALS ADVANCES 2024; 161:213884. [PMID: 38723432 DOI: 10.1016/j.bioadv.2024.213884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/05/2024] [Accepted: 04/26/2024] [Indexed: 06/04/2024]
Abstract
Prostate cancer (PCa) is a significant health problem in the male population of the Western world. Magnetic resonance elastography (MRE), an emerging medical imaging technique sensitive to mechanical properties of biological tissues, detects PCa based on abnormally high stiffness and viscosity values. Yet, the origin of these changes in tissue properties and how they correlate with histopathological markers and tumor aggressiveness are largely unknown, hindering the use of tumor biomechanical properties for establishing a noninvasive PCa staging system. To infer the contributions of extracellular matrix (ECM) components and cell motility, we investigated fresh tissue specimens from two PCa xenograft mouse models, PC3 and LNCaP, using magnetic resonance elastography (MRE), diffusion-weighted imaging (DWI), quantitative histology, and nuclear shape analysis. Increased tumor stiffness and impaired water diffusion were observed to be associated with collagen and elastin accumulation and decreased cell motility. Overall, LNCaP, while more representative of clinical PCa than PC3, accumulated fewer ECM components, induced less restriction of water diffusion, and exhibited increased cell motility, resulting in overall softer and less viscous properties. Taken together, our results suggest that prostate tumor stiffness increases with ECM accumulation and cell adhesion - characteristics that influence critical biological processes of cancer development. MRE paired with DWI provides a powerful set of imaging markers that can potentially predict prostate tumor development from benign masses to aggressive malignancies in patients. STATEMENT OF SIGNIFICANCE: Xenograft models of human prostate tumor cell lines, allowing correlation of microstructure-sensitive biophysical imaging parameters with quantitative histological methods, can be investigated to identify hallmarks of cancer.
Collapse
Affiliation(s)
- Avan Kader
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Department of Biology, Chemistry and Pharmacy, Institute of Biology, Freie Universität Berlin, Königin-Luise-Str. 1-3, 14195 Berlin, Germany; Technical University of Munich, Department of Diagnostic and Interventional Radiology, Ismaninger Str. 22, 81675 Munich, Germany.
| | - Joachim Snellings
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
| | - Lisa C Adams
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Technical University of Munich, Department of Diagnostic and Interventional Radiology, Ismaninger Str. 22, 81675 Munich, Germany.
| | - Pablo Gottheil
- Peter Debye Institute for Soft Matter Physics, Leipzig University, Linnéstraße 5, 04103 Leipzig, Germany
| | - Dilyana B Mangarova
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
| | - Jennifer L Heyl
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
| | - Jan O Kaufmann
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Bundesanstalt für Materialforschung und -prüfung (BAM), Division 1.5 Protein Analysis, Richard-Willstätter-Str. 11, 12489 Berlin, Germany.
| | - Jana Moeckel
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
| | - Julia Brangsch
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Technical University of Munich, Department of Diagnostic and Interventional Radiology, Ismaninger Str. 22, 81675 Munich, Germany.
| | - Timo A Auer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Berlin Insitute of Health (BIH), Berlin, Germany.
| | - Federico Collettini
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Berlin Insitute of Health (BIH), Berlin, Germany.
| | - Frank Sauer
- Peter Debye Institute for Soft Matter Physics, Leipzig University, Linnéstraße 5, 04103 Leipzig, Germany.
| | - Bernd Hamm
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
| | - Josef Käs
- Peter Debye Institute for Soft Matter Physics, Leipzig University, Linnéstraße 5, 04103 Leipzig, Germany.
| | - Ingolf Sack
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
| | - Marcus R Makowski
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Technical University of Munich, Department of Diagnostic and Interventional Radiology, Ismaninger Str. 22, 81675 Munich, Germany; King's College London, School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, United Kingdom.
| | - Jürgen Braun
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
9
|
Rasmussen M, Fredsøe J, Salachan PV, Blanke MPL, Larsen SH, Ulhøi BP, Jensen JB, Borre M, Sørensen KD. Stroma-specific gene expression signature identifies prostate cancer subtype with high recurrence risk. NPJ Precis Oncol 2024; 8:48. [PMID: 38395986 PMCID: PMC10891092 DOI: 10.1038/s41698-024-00540-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Current prognostic tools cannot clearly distinguish indolent and aggressive prostate cancer (PC). We hypothesized that analyzing individual contributions of epithelial and stromal components in localized PC (LPC) could improve risk stratification, as stromal subtypes may have been overlooked due to the emphasis on malignant epithelial cells. Hence, we derived molecular subtypes of PC using gene expression analysis of LPC samples from prostatectomy patients (cohort 1, n = 127) and validated these subtypes in two independent prostatectomy cohorts (cohort 2, n = 406, cohort 3, n = 126). Stroma and epithelium-specific signatures were established from laser-capture microdissection data and non-negative matrix factorization was used to identify subtypes based on these signatures. Subtypes were functionally characterized by gene set and cell type enrichment analyses, and survival analysis was conducted. Three epithelial (E1-E3) and three stromal (S1-S3) PC subtypes were identified. While subtyping based on epithelial signatures showed inconsistent associations to biochemical recurrence (BCR), subtyping by stromal signatures was significantly associated with BCR in all three cohorts, with subtype S3 indicating high BCR risk. Subtype S3 exhibited distinct features, including significantly decreased cell-polarity and myogenesis, significantly increased infiltration of M2-polarized macrophages and CD8 + T-cells compared to subtype S1. For patients clinically classified as CAPRA-S intermediate risk, S3 improved prediction of BCR. This study demonstrates the potential of stromal signatures in identification of clinically relevant PC subtypes, and further indicated that stromal characterization may enhance risk stratification in LPC and may be particularly promising in cases with high prognostic ambiguity based on clinical parameters.
Collapse
Affiliation(s)
- Martin Rasmussen
- Department of Molecular Medicine, Aarhus University Hospital (AUH), Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jacob Fredsøe
- Department of Molecular Medicine, Aarhus University Hospital (AUH), Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Paul Vinu Salachan
- Department of Molecular Medicine, Aarhus University Hospital (AUH), Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Marcus Pii Lunau Blanke
- Department of Molecular Medicine, Aarhus University Hospital (AUH), Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Stine Hesselby Larsen
- Department of Molecular Medicine, Aarhus University Hospital (AUH), Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Jørgen Bjerggaard Jensen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Urology, Gødstrup Hospital, Herning, Denmark
| | - Michael Borre
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Urology, Aarhus University Hospital (AUH), Aarhus, Denmark
| | - Karina Dalsgaard Sørensen
- Department of Molecular Medicine, Aarhus University Hospital (AUH), Aarhus, Denmark.
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
10
|
Guerrero-Ochoa P, Rodríguez-Zapater S, Anel A, Esteban LM, Camón-Fernández A, Espilez-Ortiz R, Gil-Sanz MJ, Borque-Fernando Á. Prostate Cancer and the Mevalonate Pathway. Int J Mol Sci 2024; 25:2152. [PMID: 38396837 PMCID: PMC10888820 DOI: 10.3390/ijms25042152] [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: 01/10/2024] [Revised: 02/04/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Antineoplastic therapies for prostate cancer (PCa) have traditionally centered around the androgen receptor (AR) pathway, which has demonstrated a significant role in oncogenesis. Nevertheless, it is becoming progressively apparent that therapeutic strategies must diversify their focus due to the emergence of resistance mechanisms that the tumor employs when subjected to monomolecular treatments. This review illustrates how the dysregulation of the lipid metabolic pathway constitutes a survival strategy adopted by tumors to evade eradication efforts. Integrating this aspect into oncological management could prove valuable in combating PCa.
Collapse
Affiliation(s)
- Patricia Guerrero-Ochoa
- Health Research Institute of Aragon Foundation, 50009 Zaragoza, Spain; (P.G.-O.); (A.C.-F.); (R.E.-O.); (M.J.G.-S.)
| | - Sergio Rodríguez-Zapater
- Minimally Invasive Research Group (GITMI), Faculty of Veterinary Medicine, University of Zaragoza, 50009 Zaragoza, Spain;
| | - Alberto Anel
- Department of Biochemistry and Molecular and Cellular Biology, Faculty of Sciences, University of Zaragoza, 50009 Zaragoza, Spain;
| | - Luis Mariano Esteban
- Department of Applied Mathematics, Escuela Universitaria Politécnica de La Almunia, Institute for Biocomputation and Physic of Complex Systems, Universidad de Zaragoza, 50100 La Almunia de Doña Godina, Spain
| | - Alejandro Camón-Fernández
- Health Research Institute of Aragon Foundation, 50009 Zaragoza, Spain; (P.G.-O.); (A.C.-F.); (R.E.-O.); (M.J.G.-S.)
| | - Raquel Espilez-Ortiz
- Health Research Institute of Aragon Foundation, 50009 Zaragoza, Spain; (P.G.-O.); (A.C.-F.); (R.E.-O.); (M.J.G.-S.)
- Department of Urology, Miguel Servet University Hospital, 50009 Zaragoza, Spain
- Area of Urology, Department of Surgery, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | - María Jesús Gil-Sanz
- Health Research Institute of Aragon Foundation, 50009 Zaragoza, Spain; (P.G.-O.); (A.C.-F.); (R.E.-O.); (M.J.G.-S.)
- Department of Urology, Miguel Servet University Hospital, 50009 Zaragoza, Spain
| | - Ángel Borque-Fernando
- Health Research Institute of Aragon Foundation, 50009 Zaragoza, Spain; (P.G.-O.); (A.C.-F.); (R.E.-O.); (M.J.G.-S.)
- Department of Applied Mathematics, Escuela Universitaria Politécnica de La Almunia, Institute for Biocomputation and Physic of Complex Systems, Universidad de Zaragoza, 50100 La Almunia de Doña Godina, Spain
- Department of Urology, Miguel Servet University Hospital, 50009 Zaragoza, Spain
- Area of Urology, Department of Surgery, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| |
Collapse
|
11
|
Pakula H, Omar M, Carelli R, Pederzoli F, Fanelli GN, Pannellini T, Socciarelli F, Van Emmenis L, Rodrigues S, Fidalgo-Ribeiro C, Nuzzo PV, Brady NJ, Dinalankara W, Jere M, Valencia I, Saladino C, Stone J, Unkenholz C, Garner R, Alexanderani MK, Khani F, de Almeida FN, Abate-Shen C, Greenblatt MB, Rickman DS, Barbieri CE, Robinson BD, Marchionni L, Loda M. Distinct mesenchymal cell states mediate prostate cancer progression. Nat Commun 2024; 15:363. [PMID: 38191471 PMCID: PMC10774315 DOI: 10.1038/s41467-023-44210-1] [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: 03/13/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024] Open
Abstract
In the complex tumor microenvironment (TME), mesenchymal cells are key players, yet their specific roles in prostate cancer (PCa) progression remain to be fully deciphered. This study employs single-cell RNA sequencing to delineate molecular changes in tumor stroma that influence PCa progression and metastasis. Analyzing mesenchymal cells from four genetically engineered mouse models (GEMMs) and correlating these findings with human tumors, we identify eight stromal cell populations with distinct transcriptional identities consistent across both species. Notably, stromal signatures in advanced mouse disease reflect those in human bone metastases, highlighting periostin's role in invasion and differentiation. From these insights, we derive a gene signature that predicts metastatic progression in localized disease beyond traditional Gleason scores. Our results illuminate the critical influence of stromal dynamics on PCa progression, suggesting new prognostic tools and therapeutic targets.
Collapse
Affiliation(s)
- Hubert Pakula
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Mohamed Omar
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, New York, NY, 10021, USA
| | - Ryan Carelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Filippo Pederzoli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Giuseppe Nicolò Fanelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
- Department of Laboratory Medicine, Pisa University Hospital, Division of Pathology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, 56126, Italy
| | - Tania Pannellini
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Fabio Socciarelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Lucie Van Emmenis
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Silvia Rodrigues
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Caroline Fidalgo-Ribeiro
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Pier Vitale Nuzzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Nicholas J Brady
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Wikum Dinalankara
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Madhavi Jere
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Itzel Valencia
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Christopher Saladino
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jason Stone
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Caitlin Unkenholz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Richard Garner
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Mohammad K Alexanderani
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Francesca Khani
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Francisca Nunes de Almeida
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Cory Abate-Shen
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Urology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - David S Rickman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Christopher E Barbieri
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, New York, NY, 10021, USA
- Department of Urology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Brian D Robinson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, New York, NY, 10021, USA
- Department of Urology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Luigi Marchionni
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, New York, NY, 10021, USA.
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave, Boston, MA, 02215, USA.
- University of Oxford, Nuffield Department of Surgical Sciences, Oxford, UK.
| |
Collapse
|
12
|
Sharma V, Singh A, Chauhan S, Sharma PK, Chaudhary S, Sharma A, Porwal O, Fuloria NK. Role of Artificial Intelligence in Drug Discovery and Target Identification in Cancer. Curr Drug Deliv 2024; 21:870-886. [PMID: 37670704 DOI: 10.2174/1567201821666230905090621] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/08/2023] [Accepted: 03/24/2023] [Indexed: 09/07/2023]
Abstract
Drug discovery and development (DDD) is a highly complex process that necessitates precise monitoring and extensive data analysis at each stage. Furthermore, the DDD process is both timeconsuming and costly. To tackle these concerns, artificial intelligence (AI) technology can be used, which facilitates rapid and precise analysis of extensive datasets within a limited timeframe. The pathophysiology of cancer disease is complicated and requires extensive research for novel drug discovery and development. The first stage in the process of drug discovery and development involves identifying targets. Cell structure and molecular functioning are complex due to the vast number of molecules that function constantly, performing various roles. Furthermore, scientists are continually discovering novel cellular mechanisms and molecules, expanding the range of potential targets. Accurately identifying the correct target is a crucial step in the preparation of a treatment strategy. Various forms of AI, such as machine learning, neural-based learning, deep learning, and network-based learning, are currently being utilised in applications, online services, and databases. These technologies facilitate the identification and validation of targets, ultimately contributing to the success of projects. This review focuses on the different types and subcategories of AI databases utilised in the field of drug discovery and target identification for cancer.
Collapse
Affiliation(s)
- Vishal Sharma
- Department of Pharmacy, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| | - Amit Singh
- Department of Pharmacy, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| | - Sanjana Chauhan
- Department of Pharmacy, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| | - Pramod Kumar Sharma
- Department of Pharmacy, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| | - Shubham Chaudhary
- Department of Pharmacy, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| | - Astha Sharma
- Department of Pharmacy, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| | - Omji Porwal
- Department of Pharmacognosy, Faculty of Pharmacy, Tishk International University, Erbil 44001, Iraq
| | | |
Collapse
|
13
|
An Y, Lu W, Li S, Lu X, Zhang Y, Han D, Su D, Jia J, Yuan J, Zhao B, Tu M, Li X, Wang X, Fang N, Ji S. Systematic review and integrated analysis of prognostic gene signatures for prostate cancer patients. Discov Oncol 2023; 14:234. [PMID: 38112859 PMCID: PMC10730790 DOI: 10.1007/s12672-023-00847-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023] Open
Abstract
Prostate cancer (PC) is one of the most common cancers in men and becoming the second leading cause of cancer fatalities. At present, the lack of effective strategies for prognosis of PC patients is still a problem to be solved. Therefore, it is significant to identify potential gene signatures for PC patients' prognosis. Here, we summarized 71 different prognostic gene signatures for PC and concluded 3 strategies for signature construction after extensive investigation. In addition, 14 genes frequently appeared in 71 different gene signatures, which enriched in mitotic and cell cycle. This review provides extensive understanding and integrated analysis of current prognostic signatures of PC, which may help researchers to construct gene signatures of PC and guide future clinical treatment.
Collapse
Affiliation(s)
- Yang An
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China.
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Sciences, Henan University, Jinming Street, Kaifeng, 475004, Henan, China.
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Kaifeng, 475004, China.
| | - Wenyuan Lu
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Sciences, Henan University, Jinming Street, Kaifeng, 475004, Henan, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Kaifeng, 475004, China
| | - Shijia Li
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Sciences, Henan University, Jinming Street, Kaifeng, 475004, Henan, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Kaifeng, 475004, China
| | - Xiaoyan Lu
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Sciences, Henan University, Jinming Street, Kaifeng, 475004, Henan, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Kaifeng, 475004, China
| | - Yuanyuan Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Sciences, Henan University, Jinming Street, Kaifeng, 475004, Henan, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Kaifeng, 475004, China
| | - Dongcheng Han
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Sciences, Henan University, Jinming Street, Kaifeng, 475004, Henan, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Kaifeng, 475004, China
| | - Dingyuan Su
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Sciences, Henan University, Jinming Street, Kaifeng, 475004, Henan, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Kaifeng, 475004, China
| | - Jiaxin Jia
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Sciences, Henan University, Jinming Street, Kaifeng, 475004, Henan, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Kaifeng, 475004, China
| | - Jiaxin Yuan
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Sciences, Henan University, Jinming Street, Kaifeng, 475004, Henan, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Kaifeng, 475004, China
| | - Binbin Zhao
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Sciences, Henan University, Jinming Street, Kaifeng, 475004, Henan, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Kaifeng, 475004, China
| | - Mengjie Tu
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Sciences, Henan University, Jinming Street, Kaifeng, 475004, Henan, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Kaifeng, 475004, China
| | - Xinyu Li
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Sciences, Henan University, Jinming Street, Kaifeng, 475004, Henan, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Kaifeng, 475004, China
| | - Xiaoqing Wang
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Sciences, Henan University, Jinming Street, Kaifeng, 475004, Henan, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Kaifeng, 475004, China
| | - Na Fang
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China.
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Sciences, Henan University, Jinming Street, Kaifeng, 475004, Henan, China.
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Kaifeng, 475004, China.
| | - Shaoping Ji
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China.
- Department of Biochemistry and Molecular Biology, Cell Signal Transduction Laboratory, School of Basic Medical Sciences, Henan University, Jinming Street, Kaifeng, 475004, Henan, China.
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Kaifeng, 475004, China.
| |
Collapse
|
14
|
Yuan JX, Jiang Q, Yu SJ. Diabetes mellitus and prostate cancer risk: A mendelian randomization analysis. World J Diabetes 2023; 14:1839-1848. [PMID: 38222791 PMCID: PMC10784790 DOI: 10.4239/wjd.v14.i12.1839] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/20/2023] [Accepted: 12/01/2023] [Indexed: 12/14/2023] Open
Abstract
BACKGROUND Some studies have directed towards an association between diabetes mellitus (DM) and prostate cancer (PCa); however, this specific relationship remains inconclusive. In recent years, Mendelian randomization (MR) has become a widely used analytical method for inferring epidemiological causes. AIM To investigated the potential relationship between DM and PCa using MR. METHODS We downloaded relevant data on "diabetes" and "PCa" from the IEU OpenGWAS project database, performed three different methods to conduct MR, and carried out sensitivity analysis for verification. RESULTS The results indicated that DM was an independent risk factor for PCa. The odds ratio (OR) values obtained using the inverse variance weighted method in this study were as follows: OR = 1.018 (95% confidence interval: 1.004-1.032), P = 0.014. CONCLUSION We found that DM could increase the incidence rate of PCa.
Collapse
Affiliation(s)
- Jian-Xu Yuan
- Department of Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Qing Jiang
- Department of Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Sheng-Jie Yu
- Department of Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
15
|
Logotheti S, Papadaki E, Zolota V, Logothetis C, Vrahatis AG, Soundararajan R, Tzelepi V. Lineage Plasticity and Stemness Phenotypes in Prostate Cancer: Harnessing the Power of Integrated "Omics" Approaches to Explore Measurable Metrics. Cancers (Basel) 2023; 15:4357. [PMID: 37686633 PMCID: PMC10486655 DOI: 10.3390/cancers15174357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/21/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Prostate cancer (PCa), the most frequent and second most lethal cancer type in men in developed countries, is a highly heterogeneous disease. PCa heterogeneity, therapy resistance, stemness, and lethal progression have been attributed to lineage plasticity, which refers to the ability of neoplastic cells to undergo phenotypic changes under microenvironmental pressures by switching between developmental cell states. What remains to be elucidated is how to identify measurements of lineage plasticity, how to implement them to inform preclinical and clinical research, and, further, how to classify patients and inform therapeutic strategies in the clinic. Recent research has highlighted the crucial role of next-generation sequencing technologies in identifying potential biomarkers associated with lineage plasticity. Here, we review the genomic, transcriptomic, and epigenetic events that have been described in PCa and highlight those with significance for lineage plasticity. We further focus on their relevance in PCa research and their benefits in PCa patient classification. Finally, we explore ways in which bioinformatic analyses can be used to determine lineage plasticity based on large omics analyses and algorithms that can shed light on upstream and downstream events. Most importantly, an integrated multiomics approach may soon allow for the identification of a lineage plasticity signature, which would revolutionize the molecular classification of PCa patients.
Collapse
Affiliation(s)
- Souzana Logotheti
- Department of Pathology, University of Patras, 26504 Patras, Greece; (S.L.); (E.P.); (V.Z.)
| | - Eugenia Papadaki
- Department of Pathology, University of Patras, 26504 Patras, Greece; (S.L.); (E.P.); (V.Z.)
- Department of Informatics, Ionian University, 49100 Corfu, Greece;
| | - Vasiliki Zolota
- Department of Pathology, University of Patras, 26504 Patras, Greece; (S.L.); (E.P.); (V.Z.)
| | - Christopher Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | | | - Rama Soundararajan
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vasiliki Tzelepi
- Department of Pathology, University of Patras, 26504 Patras, Greece; (S.L.); (E.P.); (V.Z.)
| |
Collapse
|
16
|
Wan B, Lu L, Lv C. Mendelian randomization study on the causal relationship between leukocyte telomere length and prostate cancer. PLoS One 2023; 18:e0286219. [PMID: 37352282 PMCID: PMC10289467 DOI: 10.1371/journal.pone.0286219] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 05/11/2023] [Indexed: 06/25/2023] Open
Abstract
BACKGROUND Leukocyte telomere length (LTL) is related to prostate cancer (PCa). However, the causal relationship between them remains unknown. This study was aimed at identifying the causal direction between LTL and PCa with Mendelian randomization (MR). METHODS Single-nucleotide polymorphisms associated with LTL were identified from a genome-wide association study (GWAS) involving 472,174 individuals. Summary-level data of PCa-related GWAS were extracted from four cohorts comprising 456,717 individuals. An inverse-variance-weighted (IVW) algorithm was used for MR. Sensitivity analyses were performed with MR-Egger regression, IVW regression, leave-one-out test, and MR-Pleiotropy Residual Sum and Outlier analyses. A meta-analysis was also performed to compute the average genetically determined effect of LTL on PCa. RESULTS A long LTL was associated with an increased risk of PCa in all cohorts, with odds ratios of 1.368 (95% confidence interval [CI]: 1.247 to 1.500, P = 2.84×10-11), 1.503 (95% CI: 1.243 to 1.816, P = 2.57×10-5), 1.722 (95% CI: 1.427 to 2.077, P = 1.48×10-8), and 1.358 (95% CI: 1.242 to 1.484, P = 1.73×10-11) in the IVW analysis. Sensitivity analyses showed that the genetically determined effect of LTL on PCa was stable and reliable. The meta-analysis showed that the genetically determined per 1-standard deviation rise in LTL correlated significantly with an average 40.6% increase in the PCa risk, with an average odds ratio of 1.406 (95% CI: 1.327 to 1.489, P < 0.001). CONCLUSION The results of this study supported the causal hypothesis that the genetically determined longer LTL was associated with a higher risk of PCa. This finding could serve as a basis for therapeutic strategies for PCa.
Collapse
Affiliation(s)
- Bangbei Wan
- Reproductive Medical Center, Hainan Women and Children’s Medical Center, Haikou, China
- Department of Urology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Likui Lu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Cai Lv
- Department of Urology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| |
Collapse
|
17
|
Lawrence MG, Taylor RA, Cuffe GB, Ang LS, Clark AK, Goode DL, Porter LH, Le Magnen C, Navone NM, Schalken JA, Wang Y, van Weerden WM, Corey E, Isaacs JT, Nelson PS, Risbridger GP. The future of patient-derived xenografts in prostate cancer research. Nat Rev Urol 2023; 20:371-384. [PMID: 36650259 PMCID: PMC10789487 DOI: 10.1038/s41585-022-00706-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2022] [Indexed: 01/19/2023]
Abstract
Patient-derived xenografts (PDXs) are generated by engrafting human tumours into mice. Serially transplantable PDXs are used to study tumour biology and test therapeutics, linking the laboratory to the clinic. Although few prostate cancer PDXs are available in large repositories, over 330 prostate cancer PDXs have been established, spanning broad clinical stages, genotypes and phenotypes. Nevertheless, more PDXs are needed to reflect patient diversity, and to study new treatments and emerging mechanisms of resistance. We can maximize the use of PDXs by exchanging models and datasets, and by depositing PDXs into biorepositories, but we must address the impediments to accessing PDXs, such as institutional, ethical and legal agreements. Through collaboration, researchers will gain greater access to PDXs representing diverse features of prostate cancer.
Collapse
Affiliation(s)
- Mitchell G Lawrence
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.
- Melbourne Urological Research Alliance, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia.
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.
- Cabrini Institute, Cabrini Health, Malvern, Victoria, Australia.
| | - Renea A Taylor
- Melbourne Urological Research Alliance, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Cabrini Institute, Cabrini Health, Malvern, Victoria, Australia
- Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Georgia B Cuffe
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Lisa S Ang
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ashlee K Clark
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- Department of Urology, Radboud University Medical Center, Nijmegen, Netherlands
| | - David L Goode
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Laura H Porter
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Clémentine Le Magnen
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Department of Urology, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Nora M Navone
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jack A Schalken
- Department of Urology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| | | | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - John T Isaacs
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter S Nelson
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Gail P Risbridger
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.
- Melbourne Urological Research Alliance, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia.
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.
- Cabrini Institute, Cabrini Health, Malvern, Victoria, Australia.
| |
Collapse
|
18
|
Pakula H, Omar M, Carelli R, Pederzoli F, Fanelli GN, Pannellini T, Van Emmenis L, Rodrigues S, Fidalgo-Ribeiro C, Nuzzo PV, Brady NJ, Jere M, Unkenholz C, Alexanderani MK, Khani F, de Almeida FN, Abate-Shen C, Greenblatt MB, Rickman DS, Barbieri CE, Robinson BD, Marchionni L, Loda M. Distinct mesenchymal cell states mediate prostate cancer progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.29.534769. [PMID: 37034687 PMCID: PMC10081210 DOI: 10.1101/2023.03.29.534769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Alterations in tumor stroma influence prostate cancer progression and metastatic potential. However, the molecular underpinnings of this stromal-epithelial crosstalk are largely unknown. Here, we compare mesenchymal cells from four genetically engineered mouse models (GEMMs) of prostate cancer representing different stages of the disease to their wild-type (WT) counterparts by single-cell RNA sequencing (scRNA-seq) and, ultimately, to human tumors with comparable genotypes. We identified 8 transcriptionally and functionally distinct stromal populations responsible for common and GEMM-specific transcriptional programs. We show that stromal responses are conserved in mouse models and human prostate cancers with the same genomic alterations. We noted striking similarities between the transcriptional profiles of the stroma of murine models of advanced disease and those of of human prostate cancer bone metastases. These profiles were then used to build a robust gene signature that can predict metastatic progression in prostate cancer patients with localized disease and is also associated with progression-free survival independent of Gleason score. Taken together, this offers new evidence that stromal microenvironment mediates prostate cancer progression, further identifying tissue-based biomarkers and potential therapeutic targets of aggressive and metastatic disease.
Collapse
Affiliation(s)
- Hubert Pakula
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Mohamed Omar
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ryan Carelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Filippo Pederzoli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Giuseppe Nicolò Fanelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Laboratory Medicine, Pisa University Hospital, Division of Pathology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56126, Italy
| | - Tania Pannellini
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Lucie Van Emmenis
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Silvia Rodrigues
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Caroline Fidalgo-Ribeiro
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Pier V. Nuzzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Nicholas J. Brady
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Madhavi Jere
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Caitlin Unkenholz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Mohammad K. Alexanderani
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Francesca Khani
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, New York, NY 10021, USA
- Department of Urology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Francisca Nunes de Almeida
- Departments of Molecular Pharmacology and Therapeutics, Urology, Medicine, Pathology & Cell Biology and Systems Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Cory Abate-Shen
- Departments of Molecular Pharmacology and Therapeutics, Urology, Medicine, Pathology & Cell Biology and Systems Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - David S. Rickman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Christopher E. Barbieri
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, New York, NY 10021, USA
- Department of Urology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Brian D. Robinson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, New York, NY 10021, USA
- Department of Urology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Luigi Marchionni
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, New York, NY 10021, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave, Boston, MA, 02215, USA
| |
Collapse
|
19
|
Chang KS, Chen ST, Sung HC, Hsu SY, Lin WY, Hou CP, Lin YH, Feng TH, Tsui KH, Juang HH. WNT1 Inducible Signaling Pathway Protein 1 Is a Stroma-Specific Secreting Protein Inducing a Fibroblast Contraction and Carcinoma Cell Growth in the Human Prostate. Int J Mol Sci 2022; 23:ijms231911437. [PMID: 36232736 PMCID: PMC9570503 DOI: 10.3390/ijms231911437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/18/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
The WNT1 inducible signaling pathway protein 1 (WISP1), a member of the connective tissue growth factor family, plays a crucial role in several important cellular functions in a highly tissue-specific manner. Results of a RT-qPCR indicated that WISP1 expressed only in cells of the human prostate fibroblasts, HPrF and WPMY-1, but not the prostate carcinoma cells in vitro. Two major isoforms (WISP1v1 and WISP1v2) were identified in the HPrF cells determined by RT-PCR and immunoblot assays. The knock-down of a WISP1 blocked cell proliferation and contraction, while treating respectively with the conditioned medium from the ectopic WISP1v1- and WISPv2-overexpressed 293T cells enhanced the migration of HPrF cells. The TNFα induced WISP1 secretion and cell contraction while the knock-down of WISP1 attenuated these effects, although TNFα did not affect the proliferation of the HPrF cells. The ectopic overexpression of WISP1v1 but not WISP1v2 downregulated the N-myc downstream regulated 1 (NDRG1) while upregulating N-cadherin, slug, snail, and vimentin gene expressions which induced not only the cell proliferation and invasion in vitro but also tumor growth of prostate carcinoma cells in vivo. The results confirmed that WISP1 is a stroma-specific secreting protein, enhancing the cell migration and contraction of prostate fibroblasts, as well as the proliferation, invasion, and tumor growth of prostate carcinoma cells.
Collapse
Affiliation(s)
- Kang-Shuo Chang
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Syue-Ting Chen
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Hsin-Ching Sung
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Shu-Yuan Hsu
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Wei-Yin Lin
- Department of Internal Medicine, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Chen-Pang Hou
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Yu-Hsiang Lin
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Tsui-Hsia Feng
- School of Nursing, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Ke-Hung Tsui
- Department of Urology, Shuang Ho Hospital, New Taipei City 235041, Taiwan
- TMU Research Center of Urology and Kidney, Department of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: (K.-H.T.); (H.-H.J.); Tel.: +886-3-2118800 (ext. 5071) (H.-H.J.); Fax: +886-3-2118112 (H.-H.J.)
| | - Horng-Heng Juang
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan
- Correspondence: (K.-H.T.); (H.-H.J.); Tel.: +886-3-2118800 (ext. 5071) (H.-H.J.); Fax: +886-3-2118112 (H.-H.J.)
| |
Collapse
|
20
|
Prostate Cancer Secretome and Membrane Proteome from Pten Conditional Knockout Mice Identify Potential Biomarkers for Disease Progression. Int J Mol Sci 2022; 23:ijms23169224. [PMID: 36012492 PMCID: PMC9409251 DOI: 10.3390/ijms23169224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is the second most common cause of mortality among men. Tumor secretome is a promising strategy for understanding the biology of tumor cells and providing markers for disease progression and patient outcomes. Here, transcriptomic-based secretome analysis was performed on the PCa tumor transcriptome of Genetically Engineered Mouse Model (GEMM) Pb-Cre4/Ptenf/f mice to identify potentially secreted and membrane proteins—PSPs and PMPs. We combined a selection of transcripts from the GSE 94574 dataset and a list of protein-coding genes of the secretome and membrane proteome datasets using the Human Protein Atlas Secretome. Notably, nine deregulated PMPs and PSPs were identified in PCa (DMPK, PLN, KCNQ5, KCNQ4, MYOC, WIF1, BMP7, F3, and MUC1). We verified the gene expression patterns of Differentially Expressed Genes (DEGs) in normal and tumoral human samples using the GEPIA tool. DMPK, KCNQ4, and WIF1 targets were downregulated in PCa samples and in the GSE dataset. A significant association between shorter survival and KCNQ4, PLN, WIF1, and F3 expression was detected in the MSKCC dataset. We further identified six validated miRNAs (mmu-miR-6962-3p, mmu-miR- 6989-3p, mmu-miR-6998-3p, mmu-miR-5627-5p, mmu-miR-15a-3p, and mmu-miR-6922-3p) interactions that target MYOC, KCNQ5, MUC1, and F3. We have characterized the PCa secretome and membrane proteome and have spotted new dysregulated target candidates in PCa.
Collapse
|
21
|
Huang S, Luo Q, Huang J, Wei J, Wang S, Hong C, Qiu P, Li C. A Cluster of Metabolic-Related Genes Serve as Potential Prognostic Biomarkers for Renal Cell Carcinoma. Front Genet 2022; 13:902064. [PMID: 35873461 PMCID: PMC9301649 DOI: 10.3389/fgene.2022.902064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/07/2022] [Indexed: 12/03/2022] Open
Abstract
Renal cell carcinoma (RCC) is the most common type of renal cancer, characterized by the dysregulation of metabolic pathways. RCC is the second highest cause of death among patients with urologic cancers and those with cancer cell metastases have a 5-years survival rate of only 10–15%. Thus, reliable prognostic biomarkers are essential tools to predict RCC patient outcomes. This study identified differentially expressed genes (DEGs) in the gene expression omnibus (GEO) database that are associated with pre-and post-metastases in clear cell renal cell carcinoma (ccRCC) patients and intersected these with metabolism-related genes in the Kyoto encyclopedia of genes and genomes (KEGG) database to identify metabolism-related DEGs (DEMGs). GOplot and ggplot packages for gene ontology (GO) and KEGG pathway enrichment analysis of DEMGs with log (foldchange) (logFC) were used to identify metabolic pathways associated with DEMG. Upregulated risk genes and downregulated protective genes among the DEMGs and seven independent metabolic genes, RRM2, MTHFD2, AGXT2, ALDH6A1, GLDC, HOGA1, and ETNK2, were found using univariate and multivariate Cox regression analysis, intersection, and Lasso-Cox regression analysis to establish a metabolic risk score signature (MRSS). Kaplan-Meier survival curve of Overall Survival (OS) showed that the low-risk group had a significantly better prognosis than the high-risk group in both the training cohort (p < 0.001; HR = 2.73, 95% CI = 1.97–3.79) and the validation cohort (p = 0.001; HR = 2.84, 95% CI = 1.50–5.38). The nomogram combined with multiple clinical information and MRSS was more effective at predicting patient outcomes than a single independent prognostic factor. The impact of metabolism on ccRCC was also assessed, and seven metabolism-related genes were established and validated as biomarkers to predict patient outcomes effectively.
Collapse
|
22
|
Dong X, Xue H, Mo F, Lin YY, Lin D, Wong NK, Sun Y, Wilkinson S, Ku AT, Hao J, Ci X, Wu R, Haegert A, Silver R, Taplin ME, Balk SP, Alumkal JJ, Sowalsky AG, Gleave M, Collins C, Wang Y. Modeling Androgen Deprivation Therapy-Induced Prostate Cancer Dormancy and Its Clinical Implications. Mol Cancer Res 2022; 20:782-793. [PMID: 35082166 PMCID: PMC9234014 DOI: 10.1158/1541-7786.mcr-21-1037] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 11/18/2022]
Abstract
Treatment-induced tumor dormancy is a state in cancer progression where residual disease is present but remains asymptomatic. Dormant cancer cells are treatment-resistant and responsible for cancer recurrence and metastasis. Prostate cancer treated with androgen-deprivation therapy (ADT) often enters a dormant state. ADT-induced prostate cancer dormancy remains poorly understood due to the challenge in acquiring clinical dormant prostate cancer cells and the lack of representative models. In this study, we aimed to develop clinically relevant models for studying ADT-induced prostate cancer dormancy. Dormant prostate cancer models were established by castrating mice bearing patient-derived xenografts (PDX) of hormonal naïve or sensitive prostate cancer. Dormancy status and tumor relapse were monitored and evaluated. Paired pre- and postcastration (dormant) PDX tissues were subjected to morphologic and transcriptome profiling analyses. As a result, we established eleven ADT-induced dormant prostate cancer models that closely mimicked the clinical courses of ADT-treated prostate cancer. We identified two ADT-induced dormancy subtypes that differed in morphology, gene expression, and relapse rates. We discovered transcriptomic differences in precastration PDXs that predisposed the dormancy response to ADT. We further developed a dormancy subtype-based, predisposed gene signature that was significantly associated with ADT response in hormonal naïve prostate cancer and clinical outcome in castration-resistant prostate cancer treated with ADT or androgen-receptor pathway inhibitors. IMPLICATIONS We have established highly clinically relevant PDXs of ADT-induced dormant prostate cancer and identified two dormancy subtypes, leading to the development of a novel predicative gene signature that allows robust risk stratification of patients with prostate cancer to ADT or androgen-receptor pathway inhibitors.
Collapse
Affiliation(s)
- Xin Dong
- Department of Experimental Therapeutics, BC Cancer Research Institute, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hui Xue
- Department of Experimental Therapeutics, BC Cancer Research Institute, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fan Mo
- Vancouver Prostate Centre, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zheijiang, China
- Hangzhou AI-Force Therapeutics, Hangzhou, Zhejiang, China
| | - Yen-yi Lin
- Vancouver Prostate Centre, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dong Lin
- Department of Experimental Therapeutics, BC Cancer Research Institute, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nelson K.Y. Wong
- Department of Experimental Therapeutics, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Yingqiang Sun
- Hangzhou AI-Force Therapeutics, Hangzhou, Zhejiang, China
| | - Scott Wilkinson
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland
| | - Anson T. Ku
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland
| | - Jun Hao
- Department of Experimental Therapeutics, BC Cancer Research Institute, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Xinpei Ci
- Department of Experimental Therapeutics, BC Cancer Research Institute, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rebecca Wu
- Department of Experimental Therapeutics, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Anne Haegert
- Vancouver Prostate Centre, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rebecca Silver
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mary-Ellen Taplin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Steven P. Balk
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Joshi J. Alumkal
- Division of Hematology and Oncology, Department of Internal Medicine, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Adam G. Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland
| | - Martin Gleave
- Vancouver Prostate Centre, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Colin Collins
- Vancouver Prostate Centre, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yuzhuo Wang
- Department of Experimental Therapeutics, BC Cancer Research Institute, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
23
|
Li R, Zhu J, Zhong W, Jia Z. Comprehensive evaluation of machine learning models and gene expression signatures for prostate cancer prognosis using large population cohorts. Cancer Res 2022; 82:1832-1843. [PMID: 35358302 DOI: 10.1158/0008-5472.can-21-3074] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 01/07/2022] [Accepted: 03/07/2022] [Indexed: 11/16/2022]
Abstract
Overtreatment remains a pervasive problem in prostate cancer (PCa) management due to the highly variable and often indolent course of disease. Molecular signatures derived from gene expression profiling have played critical roles in guiding PCa treatment decisions. Many gene expression signatures have been developed to improve the risk stratification of PCa and some of them have already been applied to clinical practice. However, no comprehensive evaluation has been performed to compare the performance of these signatures. In this study, we conducted a systematic and unbiased evaluation of 15 machine learning (ML) algorithms and 30 published PCa gene expression-based prognostic signatures leveraging 10 transcriptomics datasets with 1,558 primary PCa patients from public data repositories. This analysis revealed that survival analysis models outperformed binary classification models for risk assessment, and the performance of the survival analysis methods - Cox model regularized with ridge penalty (Cox-Ridge) and partial least squares regression for Cox model (Cox-PLS) - were generally more robust than the other methods. Based on the Cox-Ridge algorithm, several top prognostic signatures displayed comparable or even better performance than commercial panels. These findings will facilitate the identification of existing prognostic signatures that are promising for further validation in prospective studies and promote the development of robust prognostic models to guide clinical decision-making. Moreover, this study provides a valuable data resource from large primary PCa cohorts, which can be used to develop, validate, and evaluate novel statistical methodologies and molecular signatures to improve PCa management.
Collapse
Affiliation(s)
- Ruidong Li
- University of California, Riverside, Riveside, United States
| | - Jianguo Zhu
- Guizhou Provincial People's Hospital, GuiYang, Guizhou, China
| | - Weide Zhong
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhenyu Jia
- University of California of Riverside, Riverside, California, United States
| |
Collapse
|
24
|
Pan B, Wei X, Xu X. Patient-derived xenograft models in hepatopancreatobiliary cancer. Cancer Cell Int 2022; 22:41. [PMID: 35090441 PMCID: PMC8796540 DOI: 10.1186/s12935-022-02454-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 01/04/2022] [Indexed: 12/20/2022] Open
Abstract
Animal models are crucial tools for evaluating the biological progress of human cancers and for the preclinical investigation of anticancer drugs and cancer prevention. Various animals are widely used in hepatopancreatobiliary cancer research, and mouse models are the most popular. Generally, genetic tools, graft transplantation, and chemical and physical measures are adopted to generate sundry mouse models of hepatopancreatobiliary cancer. Graft transplantation is commonly used to study tumour progression. Over the past few decades, subcutaneous or orthotopic cell-derived tumour xenograft models (CDX models) have been developed to simulate distinct tumours in patients. However, two major limitations exist in CDX models. One model poorly simulates the microenvironment of tumours in humans, such as the vascular, lymphatic and immune environments. The other model loses genetic heterogeneity compared with the corresponding primary tumour. Increased efforts have focused on developing better models for hepatopancreatobiliary cancer research. Hepatopancreatobiliary cancer is considered a tumour with high molecular heterogeneity, making precision medicine challenging in cancer treatment. Developing a new animal model that can better mimic tumour tissue and more accurately predict the efficacy of anticancer treatments is urgent. For the past several years, the patient-derived xenograft model (PDX model) has emerged as a promising tool for translational research. It can retain the genetic and histological stability of their originating tumour at limited passages and shed light on precision cancer medicine. In this review, we summarize the methodology, advantages/disadvantages and applications of PDX models in hepatopancreatobiliary cancer research.
Collapse
|
25
|
Shephard AP, Giles P, Mbengue M, Alraies A, Spary LK, Kynaston H, Gurney MJ, Falcón‐Pérez JM, Royo F, Tabi Z, Parthimos D, Errington RJ, Clayton A, Webber JP. Stroma-derived extracellular vesicle mRNA signatures inform histological nature of prostate cancer. J Extracell Vesicles 2021; 10:e12150. [PMID: 34596356 PMCID: PMC8485336 DOI: 10.1002/jev2.12150] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 08/25/2021] [Accepted: 09/13/2021] [Indexed: 12/11/2022] Open
Abstract
Histological assessment of prostate cancer is the key diagnostic test and can predict disease outcome. This is however an invasive procedure that carries associated risks, hence non-invasive assays to support the diagnostic pathway are much needed. A key feature of disease progression, and subsequent poor prognosis, is the presence of an altered stroma. Here we explored the utility of prostate stromal cell-derived vesicles as indicators of an altered tumour environment. We compared vesicles from six donor-matched pairs of adjacent-normal versus disease-associated primary stromal cultures. We identified 19 differentially expressed transcripts that discriminate disease from normal stromal extracellular vesicles (EVs). EVs isolated from patient serum were investigated for these putative disease-discriminating mRNA. A set of transcripts including Caveolin-1 (CAV1), TMP2, THBS1, and CTGF were found to be successful in discriminating clinically insignificant (Gleason = 6) disease from clinically significant (Gleason > 8) prostate cancer. Furthermore, correlation between transcript expression and progression-free survival suggests that levels of these mRNA may predict disease outcome. Informed by a machine learning approach, combining measures of the five most informative EV-associated mRNAs with PSA was shown to significantly improve assay sensitivity and specificity. An in-silico model was produced, showcasing the superiority of this multi-modal liquid biopsy compared to needle biopsy for predicting disease progression. This proof of concept highlights the utility of serum EV analytics as a companion diagnostic test with prognostic utility, which may obviate the need for biopsy.
Collapse
Affiliation(s)
- Alex P. Shephard
- Tissue Microenvironment GroupDivision of Cancer and GeneticsSchool of MedicineCardiff UniversityCardiffUK
| | - Peter Giles
- Wales Gene ParkHenry Welcome BuildingCardiff UniversityCardiffUK
| | - Mariama Mbengue
- Tissue Microenvironment GroupDivision of Cancer and GeneticsSchool of MedicineCardiff UniversityCardiffUK
| | - Amr Alraies
- Tissue Microenvironment GroupDivision of Cancer and GeneticsSchool of MedicineCardiff UniversityCardiffUK
| | - Lisa K. Spary
- Wales Cancer BankUniversity Hospital of WalesCardiffUK
| | - Howard Kynaston
- Section of Surgery, Division of Cancer and Genetics, School of MedicineCardiff UniversityCardiffUK
- Department of UrologyCardiff and Vale University Health Board, University Hospital of WalesCardiffUK
| | - Mark J. Gurney
- Division of Infection and Immunity, School of MedicineCardiff UniversityCardiffUK
| | - Juan M. Falcón‐Pérez
- Exosomes Lab. CICbioGUNE‐BRTAParque TecnologicoDerioSpain
- Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas (CIBERehd)MadridSpain
- IKERBASQUEBasque Foundation for ScienceBilbaoSpain
| | - Félix Royo
- Exosomes Lab. CICbioGUNE‐BRTAParque TecnologicoDerioSpain
- Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas (CIBERehd)MadridSpain
| | - Zsuzsanna Tabi
- Tissue Microenvironment GroupDivision of Cancer and GeneticsSchool of MedicineCardiff UniversityCardiffUK
| | - Dimitris Parthimos
- Tissue Microenvironment GroupDivision of Cancer and GeneticsSchool of MedicineCardiff UniversityCardiffUK
| | - Rachel J. Errington
- Tissue Microenvironment GroupDivision of Cancer and GeneticsSchool of MedicineCardiff UniversityCardiffUK
| | - Aled Clayton
- Tissue Microenvironment GroupDivision of Cancer and GeneticsSchool of MedicineCardiff UniversityCardiffUK
| | - Jason P. Webber
- Tissue Microenvironment GroupDivision of Cancer and GeneticsSchool of MedicineCardiff UniversityCardiffUK
- Institute of Life ScienceSwansea University Medical School, Swansea UniversitySwanseaUK
| |
Collapse
|
26
|
Gumaei A, Sammouda R, Al-Rakhami M, AlSalman H, El-Zaart A. Feature selection with ensemble learning for prostate cancer diagnosis from microarray gene expression. Health Informatics J 2021; 27:1460458221989402. [PMID: 33570011 DOI: 10.1177/1460458221989402] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer diagnosis using machine learning algorithms is one of the main topics of research in computer-based medical science. Prostate cancer is considered one of the reasons that are leading to deaths worldwide. Data analysis of gene expression from microarray using machine learning and soft computing algorithms is a useful tool for detecting prostate cancer in medical diagnosis. Even though traditional machine learning methods have been successfully applied for detecting prostate cancer, the large number of attributes with a small sample size of microarray data is still a challenge that limits their ability for effective medical diagnosis. Selecting a subset of relevant features from all features and choosing an appropriate machine learning method can exploit the information of microarray data to improve the accuracy rate of detection. In this paper, we propose to use a correlation feature selection (CFS) method with random committee (RC) ensemble learning to detect prostate cancer from microarray data of gene expression. A set of experiments are conducted on a public benchmark dataset using 10-fold cross-validation technique to evaluate the proposed approach. The experimental results revealed that the proposed approach attains 95.098% accuracy, which is higher than related work methods on the same dataset.
Collapse
Affiliation(s)
- Abdu Gumaei
- Research Chair of Pervasive and Mobile Computing, King Saud University, Saudi Arabia.,Taiz University, Yemen
| | | | - Mabrook Al-Rakhami
- Research Chair of Pervasive and Mobile Computing, King Saud University, Saudi Arabia
| | | | | |
Collapse
|
27
|
Baker AT, Abuwarwar MH, Poly L, Wilkins S, Fletcher AL. Cancer-Associated Fibroblasts and T Cells: From Mechanisms to Outcomes. THE JOURNAL OF IMMUNOLOGY 2021; 206:310-320. [PMID: 33397745 DOI: 10.4049/jimmunol.2001203] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 11/06/2020] [Indexed: 12/20/2022]
Abstract
Over the past decade, T cell immunotherapy has changed the face of cancer treatment, providing robust treatment options for several previously intractable cancers. Unfortunately, many epithelial tumors with high mortality rates respond poorly to immunotherapy, and an understanding of the key impediments is urgently required. Cancer-associated fibroblasts (CAFs) comprise the most frequent nonneoplastic cellular component in most solid tumors. Far from an inert scaffold, CAFs significantly influence tumor neogenesis, persistence, and metastasis and are emerging as a key player in immunotherapy resistance. In this review, we discuss the physical and chemical barriers that CAFs place between effector T cells and their tumor cell targets, and the therapies poised to target them.
Collapse
Affiliation(s)
- Alfie T Baker
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Mohammed H Abuwarwar
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Lylarath Poly
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Simon Wilkins
- Cabrini Monash University Department of Surgery, Cabrini Hospital, Malvern 3144, Victoria, Australia.,Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne 3004, Victoria, Australia; and
| | - Anne L Fletcher
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia; .,Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| |
Collapse
|
28
|
Gevaert T, Van Eycke YR, Vanden Broeck T, Van Poppel H, Salmon I, Rorive S, Muilwijk T, Claessens F, De Ridder D, Joniau S, Decaestecker C. The potential of tumour microenvironment markers to stratify the risk of recurrence in prostate cancer patients. PLoS One 2020; 15:e0244663. [PMID: 33370412 PMCID: PMC7769484 DOI: 10.1371/journal.pone.0244663] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/15/2020] [Indexed: 12/21/2022] Open
Abstract
The tumour micro-environment (TME) plays a crucial role in the onset and progression of prostate cancer (PCa). Here we studied the potential of a selected panel of TME-markers to predict clinical recurrence (CLR) in PCa. Patient cohorts were matched for the presence or absence of CLR 5 years post-prostatectomy. Tissue micro-arrays (TMA) were composed with both prostate non-tumour (PNT) and PCa tissue and subsequently processed for immunohistochemistry (IHC). The IHC panel included markers for cancer activated fibroblasts (CAFs), blood vessels and steroid hormone receptors ((SHR): androgen receptor (AR), progesterone receptor (PR) and estrogen receptor (ER)). Stained slides were digitalised, selectively annotated and analysed for percentage of marker expression with standardized and validated image analysis algorithms. A univariable analysis identified several TME markers with significant impact on CR: expression of CD31 (vascular marker) in PNT stroma, expression of alpha smooth muscle actin (αSMA) in PCa stroma, and PR expression ratio between PCa stroma and PNT stroma. A multivariable model, which included CD31 expression (vascular marker) in PNT stroma and PR expression ratio between PCa stroma and PNT stroma, could significantly stratify patients for CLR, with the identification of a low risk and high-risk subgroup. If validated and confirmed in an independent prospective series, this subgroup might have clinical potential for PCa patient stratification.
Collapse
Affiliation(s)
- Thomas Gevaert
- Department of Urology, UZ Leuven, Leuven, Belgium
- Organ Systems, KU Leuven, Leuven, Belgium
- Department of Pathology, AZ Klina, Brasschaat, Belgium
- P.E.A.R.L. (ProstatE cAncer Research Leuven), Leuven, Belgium
| | - Yves-Rémi Van Eycke
- Laboratories of Image, Synthesis and Analysis (LISA), Brussels School of Engineering/École polytechnique de Bruxelles, ULB, Brussels, Belgium
- DIAPath-Center for Microscopy and Molecular Imaging (CMMI), ULB, Gosselies, Belgium
| | - Thomas Vanden Broeck
- Department of Urology, UZ Leuven, Leuven, Belgium
- P.E.A.R.L. (ProstatE cAncer Research Leuven), Leuven, Belgium
- Department of Molecular and Cellular Medicine, KU Leuven, Leuven, Belgium
| | - Hein Van Poppel
- Department of Urology, UZ Leuven, Leuven, Belgium
- Organ Systems, KU Leuven, Leuven, Belgium
- P.E.A.R.L. (ProstatE cAncer Research Leuven), Leuven, Belgium
| | - Isabelle Salmon
- DIAPath-Center for Microscopy and Molecular Imaging (CMMI), ULB, Gosselies, Belgium
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Centre Universitaire Inter Régional d'Expertise en Anatomie Pathologique Hospitalière (CurePath), Jumet, Belgium
| | - Sandrine Rorive
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Centre Universitaire Inter Régional d'Expertise en Anatomie Pathologique Hospitalière (CurePath), Jumet, Belgium
| | - Tim Muilwijk
- Department of Urology, UZ Leuven, Leuven, Belgium
- Organ Systems, KU Leuven, Leuven, Belgium
| | - Frank Claessens
- P.E.A.R.L. (ProstatE cAncer Research Leuven), Leuven, Belgium
- Department of Molecular and Cellular Medicine, KU Leuven, Leuven, Belgium
| | - Dirk De Ridder
- Department of Urology, UZ Leuven, Leuven, Belgium
- Organ Systems, KU Leuven, Leuven, Belgium
- P.E.A.R.L. (ProstatE cAncer Research Leuven), Leuven, Belgium
| | - Steven Joniau
- Department of Urology, UZ Leuven, Leuven, Belgium
- Organ Systems, KU Leuven, Leuven, Belgium
- P.E.A.R.L. (ProstatE cAncer Research Leuven), Leuven, Belgium
| | - Christine Decaestecker
- Laboratories of Image, Synthesis and Analysis (LISA), Brussels School of Engineering/École polytechnique de Bruxelles, ULB, Brussels, Belgium
- DIAPath-Center for Microscopy and Molecular Imaging (CMMI), ULB, Gosselies, Belgium
| |
Collapse
|
29
|
Karkampouna S, De Filippo MR, Ng CKY, Klima I, Zoni E, Spahn M, Stein F, Haberkant P, Thalmann GN, Kruithof-de Julio M. Stroma Transcriptomic and Proteomic Profile of Prostate Cancer Metastasis Xenograft Models Reveals Prognostic Value of Stroma Signatures. Cancers (Basel) 2020; 12:cancers12123786. [PMID: 33334054 PMCID: PMC7768471 DOI: 10.3390/cancers12123786] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 12/10/2020] [Indexed: 01/08/2023] Open
Abstract
Resistance acquisition to androgen deprivation treatment and metastasis progression are a major clinical issue associated with prostate cancer (PCa). The role of stroma during disease progression is insufficiently defined. Using transcriptomic and proteomic analyses on differentially aggressive patient-derived xenografts (PDXs), we investigated whether PCa tumors predispose their microenvironment (stroma) to a metastatic gene expression pattern. RNA sequencing was performed on the PCa PDXs BM18 (castration-sensitive) and LAPC9 (castration-resistant), representing different disease stages. Using organism-specific reference databases, the human-specific transcriptome (tumor) was identified and separated from the mouse-specific transcriptome (stroma). To identify proteomic changes in the tumor (human) versus the stroma (mouse), we performed human/mouse cell separation and subjected protein lysates to quantitative Tandem Mass Tag labeling and mass spectrometry. Tenascin C (TNC) was among the most abundant stromal genes, modulated by androgen levels in vivo and highly expressed in castration-resistant LAPC9 PDX. The tissue microarray of primary PCa samples (n = 210) showed that TNC is a negative prognostic marker of the clinical progression to recurrence or metastasis. Stroma markers of osteoblastic PCa bone metastases seven-up signature were induced in the stroma by the host organism in metastatic xenografts, indicating conserved mechanisms of tumor cells to induce a stromal premetastatic signature. A 50-gene list stroma signature was identified based on androgen-dependent responses, which shows a linear association with the Gleason score, metastasis progression and progression-free survival. Our data show that metastatic PCa PDXs, which differ in androgen sensitivity, trigger differential stroma responses, which show the metastasis risk stratification and prognostic biomarker potential.
Collapse
Affiliation(s)
- Sofia Karkampouna
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland; (S.K.); (M.R.D.F.); (I.K.); (E.Z.); (G.N.T.)
| | - Maria R. De Filippo
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland; (S.K.); (M.R.D.F.); (I.K.); (E.Z.); (G.N.T.)
| | - Charlotte K. Y. Ng
- Oncogenomics Laboratory, Department for BioMedical Research, University of Bern, Murtenstrasse 40, 3008 Bern, Switzerland;
| | - Irena Klima
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland; (S.K.); (M.R.D.F.); (I.K.); (E.Z.); (G.N.T.)
| | - Eugenio Zoni
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland; (S.K.); (M.R.D.F.); (I.K.); (E.Z.); (G.N.T.)
| | - Martin Spahn
- Lindenhofspital Bern, Prostate Center Bern, 3012 Bern, Switzerland;
| | - Frank Stein
- Proteomics Core Facility, EMBL Heidelberg, Meyerhofstraße 1, 69117 Heidelberg, Germany; (F.S.); (P.H.)
| | - Per Haberkant
- Proteomics Core Facility, EMBL Heidelberg, Meyerhofstraße 1, 69117 Heidelberg, Germany; (F.S.); (P.H.)
| | - George N. Thalmann
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland; (S.K.); (M.R.D.F.); (I.K.); (E.Z.); (G.N.T.)
- Department of Urology, Inselspital, Anna Seiler Haus, Bern University Hospital, 3010 Bern, Switzerland
| | - Marianna Kruithof-de Julio
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland; (S.K.); (M.R.D.F.); (I.K.); (E.Z.); (G.N.T.)
- Department of Urology, Inselspital, Anna Seiler Haus, Bern University Hospital, 3010 Bern, Switzerland
- Correspondence:
| |
Collapse
|
30
|
Chen Y, Wang W, Jiang B, Yao L, Xia F, Li X. Integrating Tumor Stroma Biomarkers With Clinical Indicators for Colon Cancer Survival Stratification. Front Med (Lausanne) 2020; 7:584747. [PMID: 33365318 PMCID: PMC7750539 DOI: 10.3389/fmed.2020.584747] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 11/12/2020] [Indexed: 01/04/2023] Open
Abstract
The tumor stroma plays an important role in tumor progression and chemotherapeutic resistance; however, its role in colon cancer (CC) survival prognosis remains to be investigated. Here, we identified tumor stroma biomarkers and evaluated their role in CC prognosis stratification. Four independent datasets containing a total of 1,313 patients were included in this study and were divided into training and testing sets. Stromal scores calculated using the estimation of stromal and immune cells in malignant tumors using expression data (ESTIMATE) algorithm were used to assess the tumor stroma level. Kaplan-Meier curves and the log-rank test were used to identify relationships between stromal score and prognosis. Tumor stroma biomarkers were identified by cross-validation of multiple datasets and bioinformatics methods. Cox proportional hazards regression models were constructed using four prognosis factors (age, tumor stage, the ESTIMATE stromal score, and the biomarker stromal score) in different combinations for prognosis prediction and compared. Patients with high stromal scores had a lower overall survival rate (p = 0.00016), higher risk of recurrence (p < 0.0001), and higher probability of chemotherapeutic resistance (p < 0.0001) than those with low scores. We identified 16 tumor stroma biomarkers and generated a new prognosis indicator termed the biomarker stromal score (ranging from 0 to 16) based on their expression levels. Its addition to an age/tumor stage-based model significantly improved prognosis prediction accuracy. In conclusion, the tumor stromal score is significantly negatively associated with CC survival prognosis, and the new tumor stroma indicator can improve CC prognosis stratification.
Collapse
Affiliation(s)
- Yong Chen
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Wenlong Wang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Bo Jiang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Yao
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Fada Xia
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xinying Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
31
|
Kato M, Sasaki T, Inoue T. Current experimental human tissue-derived models for prostate cancer research. Int J Urol 2020; 28:150-162. [PMID: 33247498 DOI: 10.1111/iju.14441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/18/2020] [Indexed: 12/17/2022]
Abstract
Scientists engaged in prostate cancer research have been conducting experiments using two-dimensional cultures of prostate cancer cell lines for decades. However, these experiments fail to reproduce and reflect the clinical course of individual patients with prostate cancer, or the molecular and genetic characteristics of prostate cancer, the basic requirement for most of the preclinical studies on prostate cancer. The use of human prostate cancer tissues in experiments has enabled the collection and verification of clinically relevant data, including chemical reactions, changes in proteins, and specific gene expression. Tissue recombination models have been employed for studying prostate development, the initiation and progression of prostate cancer, and the tumor microenvironment. Notably, the epithelial-stromal interaction, which might play a critical role in prostate cancer pathogenesis, can be reproduced in this model. Patient-derived xenograft models have been developed as powerful avatars comprising patient-derived prostate cancer tissues implanted in immunocompromised mice and could serve as a precision medicine approach for each prostate cancer patient. Spheroid and organoid assays, representative of modern three-dimensional cultures, can replicate the conditions in human prostate tumors and the prostate organ itself as a miniature model. Although an intact immune system against the tumor is missing from the models aimed at investigating immuno-oncological reagents in various malignancies, all these experimental models can help researchers in developing new drugs and selecting appropriate treatment strategies for prostate cancer patients.
Collapse
Affiliation(s)
- Manabu Kato
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Takeshi Sasaki
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Takahiro Inoue
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| |
Collapse
|
32
|
Darrell CM, Montironi R, Paner GP. Potential biomarkers and risk assessment models to enhance the tumor-node-metastasis (TNM) staging classification of urologic cancers. Expert Rev Mol Diagn 2020; 20:921-932. [PMID: 32876523 DOI: 10.1080/14737159.2020.1816827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION The anatomic-based TNM classification is considered the benchmark in cancer staging and has been regularly updated since its inception. In the current era of precision medicine, the added intention for future TNM modifications is to heighten its impact in the more 'personalized' level of cancer care. In urologic cancers, this goal may be achieved by incorporating 'non-anatomic' factors into TNM, such as biomarkers (e.g. gene alterations, molecular subtypes, genomic classifiers) and risk assessment models (e.g. nomogram, look-up table), while maintaining the anatomic extent as the foundation of staging. These different prognosticators can be combined and integrated, may serve as substratifiers for T, N, or M categories, and perhaps, incorporated as elements in TNM stage groupings to enhance their prognostic capability in urologic cancers. AREAS COVERED This review highlights candidate biomarkers and risk assessment models that can be explored to potentially improve TNM prognostication of bladder, prostate, kidney, and testicular cancers. EXPERT OPINION Recent advances in molecular analysis have increased the understanding of the genomic, transcriptomic, and epigenetic features for biomarker use in prognostication of urologic cancers, which together with the available risk assessment models, may complement and overcome the limitations of the traditional TNM staging.
Collapse
Affiliation(s)
- Caitlin M Darrell
- Departments of Pathology, Section of Urology, University of Chicago , Chicago, IL, USA
| | - Rodolfo Montironi
- School of Medicine, Section of Pathological Anatomy, Polytechnic University of the Marche Region , Ancona, Italy
| | - Gladell P Paner
- Departments of Pathology, Section of Urology, University of Chicago , Chicago, IL, USA.,Departments of Surgery, Section of Urology, University of Chicago , Chicago, IL, USA
| |
Collapse
|
33
|
Risbridger GP, Lawrence MG, Taylor RA. PDX: Moving Beyond Drug Screening to Versatile Models for Research Discovery. J Endocr Soc 2020; 4:bvaa132. [PMID: 33094211 PMCID: PMC7566391 DOI: 10.1210/jendso/bvaa132] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/10/2020] [Indexed: 01/08/2023] Open
Abstract
Patient-derived xenografts (PDXs) are tools of the trade for many researchers from all disciplines and medical specialties. Most endocrinologists, and especially those working in oncology, commonly use PDXs for preclinical drug testing and development, and over the last decade large collections of PDXs have emerged across all tumor streams. In this review, we examine how the field has evolved to include PDXs as versatile resources for research discoveries, providing evidence for guidelines and changes in clinical practice.
Collapse
Affiliation(s)
- Gail P Risbridger
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute Cancer Program, Monash University, Melbourne, Victoria, Australia.,Prostate Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Mitchell G Lawrence
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute Cancer Program, Monash University, Melbourne, Victoria, Australia.,Prostate Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Renea A Taylor
- Prostate Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,Department of Physiology, Biomedicine Discovery Institute Cancer Program, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
34
|
Linxweiler J, Hajili T, Körbel C, Berchem C, Zeuschner P, Müller A, Stöckle M, Menger MD, Junker K, Saar M. Cancer-associated fibroblasts stimulate primary tumor growth and metastatic spread in an orthotopic prostate cancer xenograft model. Sci Rep 2020; 10:12575. [PMID: 32724081 PMCID: PMC7387494 DOI: 10.1038/s41598-020-69424-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 07/03/2020] [Indexed: 12/13/2022] Open
Abstract
The unique microenvironment of the prostate plays a crucial role in the development and progression of prostate cancer (PCa). We examined the effects of cancer-associated fibroblasts (CAFs) on PCa progression using patient-derived fibroblast primary cultures in a representative orthotopic xenograft model. Primary cultures of CAFs, non-cancer-associated fibroblasts (NCAFs) and benign prostate hyperplasia-associated fibroblasts (BPHFs) were generated from patient-derived tissue specimens. These fibroblasts were coinjected together with cancer cells (LuCaP136 spheroids or LNCaP cells) in orthotopic PCa xenografts to investigate their effects on local and systemic tumor progression. Primary tumor growth as well as metastatic spread to lymph nodes and lungs were significantly stimulated by CAF coinjection in LuCaP136 xenografts. When NCAFs or BPHFs were coinjected, tumor progression was similar to injection of tumor cells alone. In LNCaP xenografts, all three fibroblast types significantly stimulated primary tumor progression compared to injection of LNCaP cells alone. CAF coinjection further increased the frequency of lymph node and lung metastases. This is the first study using an orthotopic spheroid culture xenograft model to demonstrate a stimulatory effect of patient-derived CAFs on PCa progression. The established experimental setup will provide a valuable tool to further unravel the interacting mechanisms between PCa cells and their microenvironment.
Collapse
Affiliation(s)
- Johannes Linxweiler
- Department of Urology and Pediatric Urology, Saarland University, Kirrberger Straße 100, Gebäude 6, 66424, Homburg/Saar, Germany.
| | - Turkan Hajili
- Department of Urology and Pediatric Urology, Saarland University, Kirrberger Straße 100, Gebäude 6, 66424, Homburg/Saar, Germany
| | - Christina Körbel
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Carolina Berchem
- Department of Urology and Pediatric Urology, Saarland University, Kirrberger Straße 100, Gebäude 6, 66424, Homburg/Saar, Germany
| | - Philip Zeuschner
- Department of Urology and Pediatric Urology, Saarland University, Kirrberger Straße 100, Gebäude 6, 66424, Homburg/Saar, Germany
| | - Andreas Müller
- Department of Diagnostic and Interventional Radiology, Saarland University, Homburg/Saar, Germany
| | - Michael Stöckle
- Department of Urology and Pediatric Urology, Saarland University, Kirrberger Straße 100, Gebäude 6, 66424, Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Kerstin Junker
- Department of Urology and Pediatric Urology, Saarland University, Kirrberger Straße 100, Gebäude 6, 66424, Homburg/Saar, Germany
| | - Matthias Saar
- Department of Urology and Pediatric Urology, Saarland University, Kirrberger Straße 100, Gebäude 6, 66424, Homburg/Saar, Germany
| |
Collapse
|
35
|
Luca BA, Moulton V, Ellis C, Connell SP, Brewer DS, Cooper CS. Convergence of Prognostic Gene Signatures Suggests Underlying Mechanisms of Human Prostate Cancer Progression. Genes (Basel) 2020; 11:genes11070802. [PMID: 32708551 PMCID: PMC7397325 DOI: 10.3390/genes11070802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/27/2020] [Accepted: 07/10/2020] [Indexed: 12/25/2022] Open
Abstract
The highly heterogeneous clinical course of human prostate cancer has prompted the development of multiple RNA biomarkers and diagnostic tools to predict outcome for individual patients. Biomarker discovery is often unstable with, for example, small changes in discovery dataset configuration resulting in large alterations in biomarker composition. Our hypothesis, which forms the basis of this current study, is that highly significant overlaps occurring between gene signatures obtained using entirely different approaches indicate genes fundamental for controlling cancer progression. For prostate cancer, we found two sets of signatures that had significant overlaps suggesting important genes (p < 10−34 for paired overlaps, hypergeometrical test). These overlapping signatures defined a core set of genes linking hormone signalling (HES6-AR), cell cycle progression (Prolaris) and a molecular subgroup of patients (PCS1) derived by Non Negative Matrix Factorization (NNMF) of control pathways, together designated as SIG-HES6. The second set (designated SIG-DESNT) consisted of the DESNT diagnostic signature and a second NNMF signature PCS3. Stratifications using SIG-HES6 (HES6, PCS1, Prolaris) and SIG-DESNT (DESNT) classifiers frequently detected the same individual high-risk cancers, indicating that the underlying mechanisms associated with SIG-HES6 and SIG-DESNT may act together to promote aggressive cancer development. We show that the use of combinations of a SIG-HES6 signature together with DESNT substantially increases the ability to predict poor outcome, and we propose a model for prostate cancer development involving co-operation between the SIG-HES6 and SIG-DESNT pathways that has implication for therapeutic design.
Collapse
Affiliation(s)
- Bogdan-Alexandru Luca
- School of Computing Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK; (B.-A.L.); (V.M.); (C.E.)
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK; (S.P.C.); (D.S.B.)
| | - Vincent Moulton
- School of Computing Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK; (B.-A.L.); (V.M.); (C.E.)
| | - Christopher Ellis
- School of Computing Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK; (B.-A.L.); (V.M.); (C.E.)
| | - Shea P. Connell
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK; (S.P.C.); (D.S.B.)
| | - Daniel S. Brewer
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK; (S.P.C.); (D.S.B.)
- The Earlham Institute, Norwich Research Park, Norwich NR4 7UZ, UK
| | - Colin S. Cooper
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK; (S.P.C.); (D.S.B.)
- Correspondence:
| |
Collapse
|
36
|
Bonollo F, Thalmann GN, Kruithof-de Julio M, Karkampouna S. The Role of Cancer-Associated Fibroblasts in Prostate Cancer Tumorigenesis. Cancers (Basel) 2020; 12:E1887. [PMID: 32668821 PMCID: PMC7409163 DOI: 10.3390/cancers12071887] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/10/2020] [Accepted: 07/10/2020] [Indexed: 12/16/2022] Open
Abstract
Tumors strongly depend on their surrounding tumor microenvironment (TME) for growth and progression, since stromal elements are required to generate the optimal conditions for cancer cell proliferation, invasion, and possibly metastasis. Prostate cancer (PCa), though easily curable during primary stages, represents a clinical challenge in advanced stages because of the acquisition of resistance to anti-cancer treatments, especially androgen-deprivation therapies (ADT), which possibly lead to uncurable metastases such as those affecting the bone. An increasing number of studies is giving evidence that prostate TME components, especially cancer-associated fibroblasts (CAFs), which are the most abundant cell type, play a causal role in PCa since the very early disease stages, influencing therapy resistance and metastatic progression. This is highlighted by the prognostic value of the analysis of stromal markers, which may predict disease recurrence and metastasis. However, further investigations on the molecular mechanisms of tumor-stroma interactions are still needed to develop novel therapeutic approaches targeting stromal components. In this review, we report the current knowledge of the characteristics and functions of the stroma in prostate tumorigenesis, including relevant discussion of normal prostate homeostasis, chronic inflammatory conditions, pre-neoplastic lesions, and primary and metastatic tumors. Specifically, we focus on the role of CAFs, to point out their prognostic and therapeutic potential in PCa.
Collapse
Affiliation(s)
- Francesco Bonollo
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (F.B.); (G.N.T.)
| | - George N. Thalmann
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (F.B.); (G.N.T.)
- Department of Urology, Inselspital, Bern University Hospital, 3008 Bern, Switzerland
| | - Marianna Kruithof-de Julio
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (F.B.); (G.N.T.)
- Department of Urology, Inselspital, Bern University Hospital, 3008 Bern, Switzerland
| | - Sofia Karkampouna
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (F.B.); (G.N.T.)
| |
Collapse
|
37
|
Zou MX, Zheng BW, Liu FS, Wang XB, Hu JR, Huang W, Dai ZH, Zhang QS, Liu FB, Zhong H, Jiang Y, She XL, Li XB, Lv GH, Li J. The Relationship Between Tumor-Stroma Ratio, the Immune Microenvironment, and Survival in Patients With Spinal Chordoma. Neurosurgery 2020; 85:E1095-E1110. [PMID: 31501892 DOI: 10.1093/neuros/nyz333] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 05/23/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Currently, little is known about the clinical relevance of tumor-stroma ratio (TSR) in chordoma and data discussing the relationship between TSR and immune status of chordoma are lacking. OBJECTIVE To characterize TSR distribution in spinal chordoma, and investigated its correlation with clinicopathologic or immunological features of patients and outcome. METHODS TSR was assessed visually on hematoxylin and eosin-stained sections from 54 tumor specimens by 2 independent pathologists. Multiplex immunofluorescence was used to quantify the expression levels of microvessel density, Ki-67, Brachyury, and tumor as well as stromal PD-L1. Tumor immunity status including the Immunoscore and densities of tumor-infiltrating lymphocytes (TILs) subtypes were obtained from our published data and reanalyzed. RESULTS Bland-Altman plot showed no difference between mean TSR derived from the two observers. TSR was positively associated with stromal PD-L1 expression, the Immunoscore and CD3+ as well as CD4+ TILs density, but negatively correlated with tumor microvessel density, Ki-67 index, surrounding muscle invasion by tumor and number of Foxp3+ and PD-1+ TILs. Low TSR independently predicted poor local recurrence-free survival and overall survival. Moreover, patients with low TSR and low Immunoscore chordoma phenotype were associated with the worst survival. More importantly, combined TSR and Immunoscore accurately reflected prognosis and enhanced the ability of TSR or Immunoscore alone for outcome prediction. CONCLUSION These data reveal the significant impact of TSR on tumor progression and immunological response of patients. Subsequent use of agents targeting the stroma compartment may be an effective strategy to treat chordoma especially in combination with immune-based drugs.
Collapse
Affiliation(s)
- Ming-Xiang Zou
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Bo-Wen Zheng
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Fu-Sheng Liu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Xiao-Bin Wang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Jia-Rui Hu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Wei Huang
- Institute of Precision Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Zhe-Hao Dai
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Qian-Shi Zhang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Fu-Bing Liu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Hua Zhong
- Department of Orthopedics Surgery, Central Hospital of Yi Yang, Yiyang, China
| | - Yi Jiang
- Department of Pathology, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Xiao-Ling She
- Department of Pathology, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Xiao-Bing Li
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Guo-Hua Lv
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| | - Jing Li
- Department of Spine Surgery, The Second Xiangya Hospital, Central South, University, Changsha, China
| |
Collapse
|
38
|
Oberhuber M, Pecoraro M, Rusz M, Oberhuber G, Wieselberg M, Haslinger P, Gurnhofer E, Schlederer M, Limberger T, Lagger S, Pencik J, Kodajova P, Högler S, Stockmaier G, Grund‐Gröschke S, Aberger F, Bolis M, Theurillat J, Wiebringhaus R, Weiss T, Haitel A, Brehme M, Wadsak W, Griss J, Mohr T, Hofer A, Jäger A, Pollheimer J, Egger G, Koellensperger G, Mann M, Hantusch B, Kenner L. STAT3-dependent analysis reveals PDK4 as independent predictor of recurrence in prostate cancer. Mol Syst Biol 2020; 16:e9247. [PMID: 32323921 PMCID: PMC7178451 DOI: 10.15252/msb.20199247] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 03/13/2020] [Accepted: 03/18/2020] [Indexed: 01/05/2023] Open
Abstract
Prostate cancer (PCa) has a broad spectrum of clinical behavior; hence, biomarkers are urgently needed for risk stratification. Here, we aim to find potential biomarkers for risk stratification, by utilizing a gene co-expression network of transcriptomics data in addition to laser-microdissected proteomics from human and murine prostate FFPE samples. We show up-regulation of oxidative phosphorylation (OXPHOS) in PCa on the transcriptomic level and up-regulation of the TCA cycle/OXPHOS on the proteomic level, which is inversely correlated to STAT3 expression. We hereby identify gene expression of pyruvate dehydrogenase kinase 4 (PDK4), a key regulator of the TCA cycle, as a promising independent prognostic marker in PCa. PDK4 predicts disease recurrence independent of diagnostic risk factors such as grading, staging, and PSA level. Therefore, low PDK4 is a promising marker for PCa with dismal prognosis.
Collapse
|
39
|
Alterations in the methylome of the stromal tumour microenvironment signal the presence and severity of prostate cancer. Clin Epigenetics 2020; 12:48. [PMID: 32188493 PMCID: PMC7081708 DOI: 10.1186/s13148-020-00836-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/02/2020] [Indexed: 02/07/2023] Open
Abstract
Background Prostate cancer changes the phenotype of cells within the stromal microenvironment, including fibroblasts, which in turn promote tumour progression. Functional changes in prostate cancer-associated fibroblasts (CAFs) coincide with alterations in DNA methylation levels at loci-specific regulatory regions. Yet, it is not clear how these methylation changes compare across CAFs from different patients. Therefore, we examined the consistency and prognostic significance of genome-wide DNA methylation profiles between CAFs from patients with different grades of primary prostate cancer. Results We used Infinium MethylationEPIC BeadChips to evaluate genome-wide DNA methylation profiles from 18 matched CAFs and non-malignant prostate tissue fibroblasts (NPFs) from men with moderate to high grade prostate cancer, as well as five unmatched benign prostate tissue fibroblasts (BPFs) from men with benign prostatic hyperplasia. We identified two sets of differentially methylated regions (DMRs) in patient CAFs. One set of DMRs reproducibly differed between CAFs and fibroblasts from non-malignant tissue (NPFs and BPFs). Indeed, more than 1200 DMRs consistently changed in CAFs from every patient, regardless of tumour grade. The second set of DMRs varied between CAFs according to the severity of the tumour. Notably, hypomethylation of the EDARADD promoter occurred specifically in CAFs from high-grade tumours and correlated with increased transcript abundance and increased EDARADD staining in patient tissue. Across multiple cohorts, tumours with low EDARADD DNA methylation and high EDARADD mRNA expression were consistently associated with adverse clinical features and shorter recurrence free survival. Conclusions We identified a large set of DMRs that are commonly shared across CAFs regardless of tumour grade and outcome, demonstrating highly consistent epigenome changes in the prostate tumour microenvironment. Additionally, we found that CAFs from aggressive prostate cancers have discrete methylation differences compared to CAFs from moderate risk prostate cancer. Together, our data demonstrates that the methylome of the tumour microenvironment reflects both the presence and the severity of the prostate cancer and, therefore, may provide diagnostic and prognostic potential.
Collapse
|
40
|
Sejda A, Sigorski D, Gulczyński J, Wesołowski W, Kitlińska J, Iżycka-Świeszewska E. Complexity of Neural Component of Tumor Microenvironment in Prostate Cancer. Pathobiology 2020; 87:87-99. [PMID: 32045912 DOI: 10.1159/000505437] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/16/2019] [Indexed: 11/19/2022] Open
Abstract
The tumor microenvironment (TME) plays an essential role in the development and progression of neoplasms. TME consists of the extracellular matrix and numerous specialized cells interacting with cancer cells by paracrine and autocrine mechanisms. Tumor axonogenesis and neoneurogenesis constitute a developing area of investigation. Prostate cancer (PC) is one of the most common malignancies in men worldwide. During the past years, more and more studies have shown that mechanisms leading to the development of PC are not confined only to the epithelial cancer cell, but also involve the tumor stroma. Different nerve types and neurotransmitters present within the TME are thought to be important factors in PC biology. Moreover, perineural invasion, which is a common way of PC spreading, in parallel creates the neural niche for malignant cells. Cancer neurobiology seems to have become a new discipline to explore the contribution of neoplastic cell interactions with the nervous system and the neural TME component, also to search for potential therapeutic targets in malignant tumors such as PC.
Collapse
Affiliation(s)
- Aleksandra Sejda
- Department of Pathomorphology, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland,
| | - Dawid Sigorski
- Department of Oncology, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | - Jacek Gulczyński
- Department of Pathology and Neuropathology, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Joanna Kitlińska
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Ewa Iżycka-Świeszewska
- Department of Pathology and Neuropathology, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
41
|
Nouri M, Massah S, Caradec J, Lubik AA, Li N, Truong S, Lee AR, Fazli L, Ramnarine VR, Lovnicki JM, Moore J, Wang M, Foo J, Gleave ME, Hollier BG, Nelson C, Collins C, Dong X, Buttyan R. Transient Sox9 Expression Facilitates Resistance to Androgen-Targeted Therapy in Prostate Cancer. Clin Cancer Res 2020; 26:1678-1689. [PMID: 31919137 DOI: 10.1158/1078-0432.ccr-19-0098] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 10/30/2019] [Accepted: 12/19/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Patients with metastatic prostate cancer are increasingly presenting with treatment-resistant, androgen receptor-negative/low (AR-/Low) tumors, with or without neuroendocrine characteristics, in processes attributed to tumor cell plasticity. This plasticity has been modeled by Rb1/p53 knockdown/knockout and is accompanied by overexpression of the pluripotency factor, Sox2. Here, we explore the role of the developmental transcription factor Sox9 in the process of prostate cancer therapy response and tumor progression. EXPERIMENTAL DESIGN Unique prostate cancer cell models that capture AR-/Low stem cell-like intermediates were analyzed for features of plasticity and the functional role of Sox9. Human prostate cancer xenografts and tissue microarrays were evaluated for temporal alterations in Sox9 expression. The role of NF-κB pathway activity in Sox9 overexpression was explored. RESULTS Prostate cancer stem cell-like intermediates have reduced Rb1 and p53 protein expression and overexpress Sox2 as well as Sox9. Sox9 was required for spheroid growth, and overexpression increased invasiveness and neural features of prostate cancer cells. Sox9 was transiently upregulated in castration-induced progression of prostate cancer xenografts and was specifically overexpressed in neoadjuvant hormone therapy (NHT)-treated patient tumors. High Sox9 expression in NHT-treated patients predicts biochemical recurrence. Finally, we link Sox9 induction to NF-κB dimer activation in prostate cancer cells. CONCLUSIONS Developmentally reprogrammed prostate cancer cell models recapitulate features of clinically advanced prostate tumors, including downregulated Rb1/p53 and overexpression of Sox2 with Sox9. Sox9 is a marker of a transitional state that identifies prostate cancer cells under the stress of therapeutic assault and facilitates progression to therapy resistance. Its expression may index the relative activity of the NF-κB pathway.
Collapse
Affiliation(s)
- Mannan Nouri
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada. .,Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shabnam Massah
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Josselin Caradec
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amy A Lubik
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Na Li
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sarah Truong
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Ahn R Lee
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ladan Fazli
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Varune R Ramnarine
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jessica M Lovnicki
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jackson Moore
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Mike Wang
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Jane Foo
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Martin E Gleave
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Brett G Hollier
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Colleen Nelson
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada.,Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Colin Collins
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Xuesen Dong
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ralph Buttyan
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada. .,Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
42
|
Targeting of the Cancer-Associated Fibroblast-T-Cell Axis in Solid Malignancies. J Clin Med 2019; 8:jcm8111989. [PMID: 31731701 PMCID: PMC6912330 DOI: 10.3390/jcm8111989] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/31/2019] [Accepted: 11/12/2019] [Indexed: 12/23/2022] Open
Abstract
The introduction of a wide range of immunotherapies in clinical practice has revolutionized the treatment of cancer in the last decade. The majority of these therapeutic modalities are centered on reinvigorating a tumor-reactive cytotoxic T-cell response. While impressive clinical successes are obtained, the majority of cancer patients still fail to show a clinical response, despite the fact that their tumors express antigens that can be recognized by the immune system. This is due to a series of other cellular actors, present in or attracted towards the tumor microenvironment, including regulatory T-cells, myeloid-derived suppressor cells and cancer-associated fibroblasts (CAFs). As the main cellular constituent of the tumor-associated stroma, CAFs form a heterogeneous group of cells which can drive cancer cell invasion but can also impair the migration and activation of T-cells through direct and indirect mechanisms. This singles CAFs out as an important next target for further optimization of T-cell based immunotherapies. Here, we review the recent literature on the role of CAFs in orchestrating T-cell activation and migration within the tumor microenvironment and discuss potential avenues for targeting the interactions between fibroblasts and T-cells.
Collapse
|
43
|
Blom S, Erickson A, Östman A, Rannikko A, Mirtti T, Kallioniemi O, Pellinen T. Fibroblast as a critical stromal cell type determining prognosis in prostate cancer. Prostate 2019; 79:1505-1513. [PMID: 31269283 PMCID: PMC6813917 DOI: 10.1002/pros.23867] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Tumor stroma associates with prostate cancer (PCa) progression, but its specific cellular composition and association to patient survival outcome have not been characterized. METHODS We analyzed stromal composition in human PCa using multiplex immunohistochemistry and quantitative, high-resolution image analysis in two retrospective, formalin-fixed paraffin embedded observational clinical cohorts (Cohort I, n = 117; Cohort II, n = 340) using PCa-specific mortality as outcome measurement. RESULTS A high proportion of fibroblasts associated with aggressive disease and castration-resistant prostate cancer (CRPC). In a multivariate analysis, increase in fibroblast proportion predicted poor cancer-specific outcome independently in the two clinical cohorts studied. CONCLUSIONS Fibroblasts were the most important cell type in determining prognosis in PCa and associated with CRPC. Thus, the stromal composition could be critically important in developing diagnostic and therapeutic approaches to aggressive prostate cancer.
Collapse
Affiliation(s)
- Sami Blom
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE)University of HelsinkiHelsinkiFinland
| | - Andrew Erickson
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE)University of HelsinkiHelsinkiFinland
| | - Arne Östman
- Science for Life Laboratory, Department of Oncology and PathologyKarolinska InstitutetStockholmSweden
| | - Antti Rannikko
- Department of UrologyHelsinki University and Helsinki University HospitalHelsinkiFinland
| | - Tuomas Mirtti
- Department of PathologyUniversity of HelsinkiHelsinkiFinland
| | - Olli Kallioniemi
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE)University of HelsinkiHelsinkiFinland
- Science for Life Laboratory, Department of Oncology and PathologyKarolinska InstitutetStockholmSweden
| | - Teijo Pellinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE)University of HelsinkiHelsinkiFinland
| |
Collapse
|
44
|
Boufaied N, Takhar M, Nash C, Erho N, Bismar TA, Davicioni E, Thomson AA. Development of a predictive model for stromal content in prostate cancer samples to improve signature performance. J Pathol 2019; 249:411-424. [PMID: 31206668 PMCID: PMC6900085 DOI: 10.1002/path.5315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 05/27/2019] [Accepted: 06/13/2019] [Indexed: 01/23/2023]
Abstract
Prostate cancer is heterogeneous in both cellular composition and patient outcome, and development of biomarker signatures to distinguish indolent from aggressive tumours is a high priority. Stroma plays an important role during prostate cancer progression and undergoes histological and transcriptional changes associated with disease. However, identification and validation of stromal markers is limited by a lack of datasets with defined stromal/tumour ratio. We have developed a prostate‐selective signature to estimate the stromal content in cancer samples of mixed cellular composition. We identified stromal‐specific markers from transcriptomic datasets of developmental prostate mesenchyme and prostate cancer stroma. These were experimentally validated in cell lines, datasets of known stromal content, and by immunohistochemistry in tissue samples to verify stromal‐specific expression. Linear models based upon six transcripts were able to infer the stromal content and estimate stromal composition in mixed tissues. The best model had a coefficient of determination R2 of 0.67. Application of our stromal content estimation model in various prostate cancer datasets led to improved performance of stromal predictive signatures for disease progression and metastasis. The stromal content of prostate tumours varies considerably; consequently, deconvolution of stromal proportion may yield better results than tumour cell deconvolution. We suggest that adjusting expression data for cell composition will improve stromal signature performance and lead to better prognosis and stratification of men with prostate cancer. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Nadia Boufaied
- Division of Urology and Cancer Research Program, McGill University Health Centre Research Institute, Quebec, Canada
| | - Mandeep Takhar
- Research and Development, GenomeDx Biosciences, Vancouver, Canada
| | - Claire Nash
- Division of Urology and Cancer Research Program, McGill University Health Centre Research Institute, Quebec, Canada
| | - Nicholas Erho
- Research and Development, GenomeDx Biosciences, Vancouver, Canada
| | - Tarek A Bismar
- Department of Pathology and Laboratory Medicine, University of Calgary Cumming School of Medicine, Calgary, Canada.,Department of Oncology, Biochemistry and Molecular Biology, University of Calgary Cumming School of Medicine, Calgary, Canada
| | - Elai Davicioni
- Research and Development, GenomeDx Biosciences, Vancouver, Canada
| | - Axel A Thomson
- Division of Urology and Cancer Research Program, McGill University Health Centre Research Institute, Quebec, Canada
| |
Collapse
|
45
|
Frame FM, Maitland NJ. Epigenetic Control of Gene Expression in the Normal and Malignant Human Prostate: A Rapid Response Which Promotes Therapeutic Resistance. Int J Mol Sci 2019; 20:E2437. [PMID: 31108832 PMCID: PMC6566891 DOI: 10.3390/ijms20102437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/24/2019] [Accepted: 04/29/2019] [Indexed: 12/12/2022] Open
Abstract
A successful prostate cancer must be capable of changing its phenotype in response to a variety of microenvironmental influences, such as adaptation to treatment or successful proliferation at a particular metastatic site. New cell phenotypes emerge by selection from the large, genotypically heterogeneous pool of candidate cells present within any tumor mass, including a distinct stem cell-like population. In such a multicellular model of human prostate cancer, flexible responses are primarily governed not only by de novo mutations but appear to be dominated by a combination of epigenetic controls, whose application results in treatment resistance and tumor relapse. Detailed studies of these individual cell populations have resulted in an epigenetic model for epithelial cell differentiation, which is also instructive in explaining the reported high and inevitable relapse rates of human prostate cancers to a multitude of treatment types.
Collapse
Affiliation(s)
- Fiona M Frame
- The Cancer Research Unit, Department of Biology, University of York, Heslington, York YO10 5DD, UK.
| | - Norman J Maitland
- The Cancer Research Unit, Department of Biology, University of York, Heslington, York YO10 5DD, UK.
| |
Collapse
|
46
|
Sacca PA, Mazza ON, Scorticati C, Vitagliano G, Casas G, Calvo JC. Human Periprostatic Adipose Tissue: Secretome from Patients With Prostate Cancer or Benign Prostate Hyperplasia. Cancer Genomics Proteomics 2019; 16:29-58. [PMID: 30587498 DOI: 10.21873/cgp.20110] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 10/10/2018] [Accepted: 10/12/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND/AIM Periprostatic adipose tissue (PPAT) directs tumour behaviour. Microenvironment secretome provides information related to its biology. This study was performed to identify secreted proteins by PPAT, from both prostate cancer and benign prostate hyperplasia (BPH) patients. PATIENTS AND METHODS Liquid chromatography-mass spectrometry-based proteomic analysis was performed in PPAT-conditioned media (CM) from patients with prostate cancer (CMs-T) (stage T3: CM-T3, stage T2: CM-T2) or benign disease (CM-BPH). RESULTS The highest number and diversity of proteins was identified in CM-T3. Locomotion was the biological process mainly associated to CMs-T and reproduction to CM-T3. Immune responses were enriched in CMs-T. Extracellular matrix and structural proteins were associated to CMs-T. CM-T3 was enriched in proteins with catalytic activity and CM-T2 in proteins with defense/immunity activity. Metabolism and energy pathways were enriched in CM-T3 and those with immune system functions in CMs-T. Transport proteins were enriched in CM-T2 and CM-BPH. CONCLUSION Proteins and pathways reported in this study could be useful to distinguish stages of disease and may become targets for novel therapies.
Collapse
Affiliation(s)
- Paula Alejandra Sacca
- Institute of Biology and Experimental Medicine (IBYME), CONICET, Buenos Aires, Argentina
| | - Osvaldo Néstor Mazza
- Department of Urology, School of Medicine, University of Buenos Aires, Clínical Hospital "José de San Martín", Buenos Aires, Argentina
| | - Carlos Scorticati
- Department of Urology, School of Medicine, University of Buenos Aires, Clínical Hospital "José de San Martín", Buenos Aires, Argentina
| | | | - Gabriel Casas
- Department of Pathology, Deutsches Hospital, Buenos Aires, Argentina
| | - Juan Carlos Calvo
- Institute of Biology and Experimental Medicine (IBYME), CONICET, Buenos Aires, Argentina.,Department of Biological Chemistry, School of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
47
|
Lu H, Arshad M, Thornton A, Avesani G, Cunnea P, Curry E, Kanavati F, Liang J, Nixon K, Williams ST, Hassan MA, Bowtell DDL, Gabra H, Fotopoulou C, Rockall A, Aboagye EO. A mathematical-descriptor of tumor-mesoscopic-structure from computed-tomography images annotates prognostic- and molecular-phenotypes of epithelial ovarian cancer. Nat Commun 2019; 10:764. [PMID: 30770825 PMCID: PMC6377605 DOI: 10.1038/s41467-019-08718-9] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 01/24/2019] [Indexed: 12/11/2022] Open
Abstract
The five-year survival rate of epithelial ovarian cancer (EOC) is approximately 35-40% despite maximal treatment efforts, highlighting a need for stratification biomarkers for personalized treatment. Here we extract 657 quantitative mathematical descriptors from the preoperative CT images of 364 EOC patients at their initial presentation. Using machine learning, we derive a non-invasive summary-statistic of the primary ovarian tumor based on 4 descriptors, which we name "Radiomic Prognostic Vector" (RPV). RPV reliably identifies the 5% of patients with median overall survival less than 2 years, significantly improves established prognostic methods, and is validated in two independent, multi-center cohorts. Furthermore, genetic, transcriptomic and proteomic analysis from two independent datasets elucidate that stromal phenotype and DNA damage response pathways are activated in RPV-stratified tumors. RPV and its associated analysis platform could be exploited to guide personalized therapy of EOC and is potentially transferrable to other cancer types.
Collapse
Affiliation(s)
- Haonan Lu
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Mubarik Arshad
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Andrew Thornton
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Giacomo Avesani
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Paula Cunnea
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Ed Curry
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Fahdi Kanavati
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Jack Liang
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Katherine Nixon
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Sophie T Williams
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Mona Ali Hassan
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - David D L Bowtell
- Peter MacCallum Cancer Centre, Melbourne, 3010, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, 3010, VIC, Australia
| | - Hani Gabra
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
- Early Clinical Development, iMED Biotech Unit, AstraZeneca, Cambridge, SG8 6HB, UK
| | - Christina Fotopoulou
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Andrea Rockall
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
- Department of Radiology, Imperial College Healthcare NHS Trust, London, W12 0HS, UK
- Department of Radiology, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Eric O Aboagye
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK.
| |
Collapse
|
48
|
Namekawa T, Ikeda K, Horie-Inoue K, Inoue S. Application of Prostate Cancer Models for Preclinical Study: Advantages and Limitations of Cell Lines, Patient-Derived Xenografts, and Three-Dimensional Culture of Patient-Derived Cells. Cells 2019; 8:cells8010074. [PMID: 30669516 PMCID: PMC6357050 DOI: 10.3390/cells8010074] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 01/17/2019] [Accepted: 01/18/2019] [Indexed: 12/13/2022] Open
Abstract
Various preclinical models have been developed to clarify the pathophysiology of prostate cancer (PCa). Traditional PCa cell lines from clinical metastatic lesions, as exemplified by DU-145, PC-3, and LNCaP cells, are useful tools to define mechanisms underlying tumorigenesis and drug resistance. Cell line-based experiments, however, have limitations for preclinical studies because those cells are basically adapted to 2-dimensional monolayer culture conditions, in which the majority of primary PCa cells cannot survive. Recent tissue engineering enables generation of PCa patient-derived xenografts (PDXs) from both primary and metastatic lesions. Compared with fresh PCa tissue transplantation in athymic mice, co-injection of PCa tissues with extracellular matrix in highly immunodeficient mice has remarkably improved the success rate of PDX generation. PDX models have advantages to appropriately recapitulate the molecular diversity, cellular heterogeneity, and histology of original patient tumors. In contrast to PDX models, patient-derived organoid and spheroid PCa models in 3-dimensional culture are more feasible tools for in vitro studies for retaining the characteristics of patient tumors. In this article, we review PCa preclinical model cell lines and their sublines, PDXs, and patient-derived organoid and spheroid models. These PCa models will be applied to the development of new strategies for cancer precision medicine.
Collapse
Affiliation(s)
- Takeshi Namekawa
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama 350-1241, Japan.
- Department of Urology, Graduate School of Medicine, Chiba University, Chiba, Chiba 260-8677, Japan.
| | - Kazuhiro Ikeda
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama 350-1241, Japan.
| | - Kuniko Horie-Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama 350-1241, Japan.
| | - Satoshi Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama 350-1241, Japan.
- Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Itabashi-ku, Tokyo 173-0015, Japan.
| |
Collapse
|
49
|
Lange T, Oh-Hohenhorst SJ, Joosse SA, Pantel K, Hahn O, Gosau T, Dyshlovoy SA, Wellbrock J, Feldhaus S, Maar H, Gehrcke R, Kluth M, Simon R, Schlomm T, Huland H, Schumacher U. Development and Characterization of a Spontaneously Metastatic Patient-Derived Xenograft Model of Human Prostate Cancer. Sci Rep 2018; 8:17535. [PMID: 30510249 PMCID: PMC6277427 DOI: 10.1038/s41598-018-35695-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/09/2018] [Indexed: 12/15/2022] Open
Abstract
Here we describe the establishment and characterization of an AR+, PSMA+, ERG+, PTEN-/-, CHD1+/- patient-derived xenograft (PDX) model termed 'C5', which has been developed from a 60 years old patient suffering from castration-resistant prostate cancer (CRPC). The patient underwent radical prostatectomy, showed early tumor marker PSA recurrence and, one year after surgery, abiraterone resistance. Subcutaneous C5 tumors can be serially transplanted between mice and grow within ~90 days to 1.5-2 cm³ tumors in SCID Balb/c mice (take rate 100%), NOD-scid IL2Rgnull (NSG) mice (100%) and C57BL/6 pfp-/-/rag2-/- mice (66%). In contrast, no tumor growth is observed in female mice. C5 tumors can be cryopreserved and show the same growth characteristics in vivo afterwards. C5 tumor cells do not grow stably in vitro, neither under two- nor three-dimensional cell culture conditions. Upon serial transplantation, some C5 tumors spontaneously disseminated to distant sites with an observable trend towards higher metastatic cell loads in scid compared to NSG mice. Lung metastases could be verified by histology by means of anti-PSMA immunohistochemistry, exclusively demonstrating single disseminated tumor cells (DTCs) and micro-metastases. Upon surgical resection of the primary tumors, such pulmonary foci rarely grew out to multi-cellular metastatic colonies despite doubled overall survival span. In the brain and bone marrow, the metastatic cell load present at surgery even disappeared during the post-surgical period. We provide shallow whole genome sequencing and whole exome sequencing data of C5 tumors demonstrating the copy number aberration/ mutation status of this PCa model and proving genomic stability over several passages. Moreover, we analyzed genomic and transcriptomic alterations during metastatic progression achieved by serial transplantation. This study describes a novel PCa PDX model that enables future research on several aspects of metastatic PCa, particularly for the AR+ , ERG+ , PTEN-/- PCa subtype.
Collapse
Affiliation(s)
- Tobias Lange
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| | - Su Jung Oh-Hohenhorst
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon A Joosse
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Oliver Hahn
- Department of Urology, University Medical Center Goettingen, Robert-Koch-Strasse 40, 37075, Goettingen, Germany
| | - Tobias Gosau
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Sergey A Dyshlovoy
- Laboratory of Experimental Oncology, Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,School of Natural Sciences, Far Eastern Federal University, Vladivostok, Russian Federation
| | - Jasmin Wellbrock
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Susanne Feldhaus
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Hanna Maar
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Renate Gehrcke
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schlomm
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Urology, Charité University Hospital, Berlin, Germany
| | - Hartwig Huland
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| |
Collapse
|
50
|
Schmidt L, Møller M, Haldrup C, Strand SH, Vang S, Hedegaard J, Høyer S, Borre M, Ørntoft T, Sørensen KD. Exploring the transcriptome of hormone-naive multifocal prostate cancer and matched lymph node metastases. Br J Cancer 2018; 119:1527-1537. [PMID: 30449885 PMCID: PMC6288156 DOI: 10.1038/s41416-018-0321-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 10/09/2018] [Accepted: 10/09/2018] [Indexed: 11/09/2022] Open
Abstract
Background The current inability to predict whether a primary prostate cancer (PC) will progress to metastatic disease leads to overtreatment of indolent PCs as well as undertreatment of aggressive PCs. Here, we explored the transcriptional changes associated with metastatic progression of multifocal hormone-naive PC. Methods Using total RNA-sequencing, we analysed laser micro-dissected primary PC foci (n = 23), adjacent normal prostate tissue samples (n = 23) and lymph node metastases (n = 9) from ten hormone-naive PC patients. Genes important for PC progression were identified using differential gene expression and clustering analysis. From these, two multi-gene-based expression signatures (models) were developed, and their prognostic potential was evaluated using Cox-regression and Kaplan–Meier analyses in three independent radical prostatectomy (RP) cohorts (>650 patients). Results We identified several novel PC-associated transcripts deregulated during PC progression, and these transcripts were used to develop two novel gene-expression-based prognostic models. The models showed independent prognostic potential in three RP cohorts (n = 405, n = 107 and n = 91), using biochemical recurrence after RP as the primary clinical endpoint. Conclusions We identified several transcripts deregulated during PC progression and developed two new prognostic models for PC risk stratification, each of which showed independent prognostic value beyond routine clinicopathological factors in three independent RP cohorts.
Collapse
Affiliation(s)
- Linnéa Schmidt
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Mia Møller
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Christa Haldrup
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Siri H Strand
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Søren Vang
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Jakob Hedegaard
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Søren Høyer
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Michael Borre
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Torben Ørntoft
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|