1
|
Liang H, Zhou B, Li P, Zhang X, Zhang S, Zhang Y, Yao S, Qu S, Chen J. Stemness regulation in prostate cancer: prostate cancer stem cells and targeted therapy. Ann Med 2025; 57:2442067. [PMID: 39711287 DOI: 10.1080/07853890.2024.2442067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Increasing evidence indicates that cancer stem cells (CSCs) and cancer stem-like cells form a special subpopulation of cells that are ubiquitous in tumors. These cells exhibit similar characteristics to those of normal stem cells in tissues; moreover, they are capable of self-renewal and differentiation, as well as high tumorigenicity and drug resistance. In prostate cancer (PCa), it is difficult to kill these cells using androgen signaling inhibitors and chemotherapy drugs. Consequently, the residual prostate cancer stem cells (PCSCs) mediate tumor recurrence and progression. OBJECTIVE This review aims to provide a comprehensive and up-to-date overview of PCSCs, with a particular emphasis on potential therapeutic strategies targeting these cells. METHODS After searching in PubMed and Embase databases using 'prostate cancer' and 'cancer stem cells' as keywords, studies related were compiled and examined. RESULTS In this review, we detail the origin and characteristics of PCSCs, introduce the regulatory pathways closely related to CSC survival and stemness maintenance, and discuss the link between epithelial-mesenchymal transition, tumor microenvironment and tumor stemness. Furthermore, we introduce the currently available therapeutic strategies targeting CSCs, including signaling pathway inhibitors, anti-apoptotic protein inhibitors, microRNAs, nanomedicine, and immunotherapy. Lastly, we summarize the limitations of current CSC research and mention future research directions. CONCLUSION A deeper understanding of the regulatory network and molecular markers of PCSCs could facilitate the development of novel therapeutic strategies targeting these cells. Previous preclinical studies have demonstrated the potential of this treatment approach. In the future, this may offer alternative treatment options for PCa patients.
Collapse
Affiliation(s)
- Hao Liang
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Bin Zhou
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Peixin Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoyi Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shijie Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Yaozhong Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shengwen Yao
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Sifeng Qu
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Jun Chen
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| |
Collapse
|
2
|
Liu S, Nam HS, Zeng Z, Deng X, Pashaei E, Zang Y, Yang L, Li C, Huang J, Wendt MK, Lu X, Huang R, Wan J. CDHu40: a novel marker gene set of neuroendocrine prostate cancer. Brief Bioinform 2024; 25:bbae471. [PMID: 39318189 PMCID: PMC11422505 DOI: 10.1093/bib/bbae471] [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: 04/01/2024] [Revised: 07/22/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024] Open
Abstract
Prostate cancer (PCa) is the most prevalent cancer affecting American men. Castration-resistant prostate cancer (CRPC) can emerge during hormone therapy for PCa, manifesting with elevated serum prostate-specific antigen levels, continued disease progression, and/or metastasis to the new sites, resulting in a poor prognosis. A subset of CRPC patients shows a neuroendocrine (NE) phenotype, signifying reduced or no reliance on androgen receptor signaling and a particularly unfavorable prognosis. In this study, we incorporated computational approaches based on both gene expression profiles and protein-protein interaction networks. We identified 500 potential marker genes, which are significantly enriched in cell cycle and neuronal processes. The top 40 candidates, collectively named CDHu40, demonstrated superior performance in distinguishing NE PCa (NEPC) and non-NEPC samples based on gene expression profiles. CDHu40 outperformed most of the other published marker sets, excelling particularly at the prognostic level. Notably, some marker genes in CDHu40, absent in the other marker sets, have been reported to be associated with NEPC in the literature, such as DDC, FOLH1, BEX1, MAST1, and CACNA1A. Importantly, elevated CDHu40 scores derived from our predictive model showed a robust correlation with unfavorable survival outcomes in patients, indicating the potential of the CDHu40 score as a promising indicator for predicting the survival prognosis of those patients with the NE phenotype. Motif enrichment analysis on the top candidates suggests that REST and E2F6 may serve as key regulators in the NEPC progression.
Collapse
Affiliation(s)
- Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, 410 W 10th Street, Indianapolis, IN 46202, United States
| | - Hye Seung Nam
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, United States
| | - Ziyu Zeng
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, 100 Galvin Life Science Center, Notre Dame, IN 46556, United States
| | - Xuehong Deng
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, United States
| | - Elnaz Pashaei
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, 410 W 10th Street, Indianapolis, IN 46202, United States
| | - Yong Zang
- Department of Biostatistics & Health Data Science, Indiana University School of Medicine, 410 W 10th Street, Indianapolis, IN 46202, United States
| | - Lei Yang
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W Walnut St, Indianapolis, IN 46202, United States
| | - Chenglong Li
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 1345 Center Dr Room P3-12, Gainesville, FL 32603, United States
| | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine, Davison Building, 40 Duke Medicine, Durham, NC 27710, United States
| | - Michael K Wendt
- Department of Internal Medicine, Division of Hematology and Oncology, University of Iowa, 200 Hawkins Dr, Iowa City, IA 52242, United States
- Holden Comprehensive Cancer Center, University of Iowa, 200 Hawkins Dr, Iowa City, IA, 52242, United States
| | - Xin Lu
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, 100 Galvin Life Science Center, Notre Dame, IN 46556, United States
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, 535 Barnhill Dr, Indianapolis, IN 46202, United States
| | - Rong Huang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, United States
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, 410 W 10th Street, Indianapolis, IN 46202, United States
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, 535 Barnhill Dr, Indianapolis, IN 46202, United States
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, 410 W 10th Street, Indianapolis, IN 46202, United States
| |
Collapse
|
3
|
Su H, Huang L, Zhou J, Yang G. Prostate cancer stem cells and their targeted therapies. Front Cell Dev Biol 2024; 12:1410102. [PMID: 39175878 PMCID: PMC11338935 DOI: 10.3389/fcell.2024.1410102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 07/23/2024] [Indexed: 08/24/2024] Open
Abstract
Prostate cancer (PCa) is the most common malignancy among men worldwide. Through androgen receptor signaling inhibitor (ARSI) treatment, patients eventually succumb to castration-resistant prostate cancer (CRPC). For this, the prostate cancer stem cells (PCSCs), as a minor population of tumor cells that can promote tumor relapse, ARSI resistance, and disease progression, are gaining attention. Therefore, specific therapy targeting PCSCs has momentum. This study reviewed the identification and characterization of PCSCs and PCSC-based putative biomarkers and summarized their mechanisms of action. We further discussed clinical trials of novel therapeutic interventions focused on PCSC-related pathways, the PCSC microenvironment, cutting-edge miRNA therapy, and immunotherapy approaches from a mechanistic standpoint. This review provides updated insights into PCSC plasticity, identifying new PCSC biomarkers and optimized treatments for patients with advanced PCa.
Collapse
Affiliation(s)
- Huilan Su
- Research Center for Translational Medicine, Cancer Stem Cell Institute, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liqun Huang
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianjun Zhou
- Research Center for Translational Medicine, Cancer Stem Cell Institute, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guosheng Yang
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Li L, Hyun Cho K, Yu X, Cheng S. Systematic Multi-Omics Investigation of Androgen Receptor Driven Gene Expression and Epigenetics changes in Prostate Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.22.604505. [PMID: 39091838 PMCID: PMC11291036 DOI: 10.1101/2024.07.22.604505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Background Prostate cancer, a common malignancy, is driven by androgen receptor (AR) signaling. Understanding the function of AR signaling is critical for prostate cancer research. Methods We performed multi-omics data analysis for the AR+, androgen-sensitive LNCaP cell line, focusing on gene expression (RNAseq), chromatin accessibility (ATACseq), and transcription factor binding (ChIPseq). High-quality datasets were curated from public repositories and processed using state-of-the-art bioinformatics tools. Results Our analysis identified 1004 up-regulated and 707 down-regulated genes in response to androgen deprivation therapy (ADT) which diminished AR signaling activity. Gene-set enrichment analysis revealed that AR signaling influences pathways related to neuron differentiation, cell adhesion, P53 signaling, and inflammation. ATACseq and ChIPseq data demonstrated that as a transcription factor, AR primarily binds to distal enhancers, influencing chromatin modifications without affecting proximal promoter regions. In addition, the AR-induced genes maintained higher active chromatin states than AR-inhibited genes, even under ADT conditions. Furthermore, ADT did not directly induce neuroendocrine differentiation in LNCaP cells, suggesting a complex mechanism behind neuroendocrine prostate cancer development. In addition, a publicly available online application LNCaP-ADT (https://pcatools.shinyapps.io/shinyADT/) was launched for users to visualize and browse data generated by this study. Conclusion This study provides a comprehensive multi-omics dataset, elucidating the role of AR signaling in prostate cancer at the transcriptomic and epigenomic levels. The reprocessed data is publicly available, offering a valuable resource for future prostate cancer research.
Collapse
Affiliation(s)
- Lin Li
- Department of Biochemistry and Molecular biology, LSU Health Shreveport, Shreveport, LA
- Feist-Weiller Cancer Center, LSU Health Shreveport, Shreveport, LA
| | - Kyung Hyun Cho
- Department of Biochemistry and Molecular biology, LSU Health Shreveport, Shreveport, LA
| | - Xiuping Yu
- Department of Biochemistry and Molecular biology, LSU Health Shreveport, Shreveport, LA
- Feist-Weiller Cancer Center, LSU Health Shreveport, Shreveport, LA
- Department of Urology, LSU Health Shreveport, Shreveport, LA
| | - Siyuan Cheng
- Department of Biochemistry and Molecular biology, LSU Health Shreveport, Shreveport, LA
- Feist-Weiller Cancer Center, LSU Health Shreveport, Shreveport, LA
| |
Collapse
|
5
|
Uo T, Ojo KK, Sprenger CC, Soriano Epilepsia K, Perera BGK, Damodarasamy M, Sun S, Kim S, Hogan HH, Hulverson MA, Choi R, Whitman GR, Barrett LK, Michaels SA, Xu LH, Sun VL, Arnold SL, Pang HJ, Nguyen MM, Vigil ALB, Kamat V, Sullivan LB, Sweet IR, Vidadala R, Maly DJ, Van Voorhis WC, Plymate SR. A Compound That Inhibits Glycolysis in Prostate Cancer Controls Growth of Advanced Prostate Cancer. Mol Cancer Ther 2024; 23:973-994. [PMID: 38507737 PMCID: PMC11219269 DOI: 10.1158/1535-7163.mct-23-0540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/19/2023] [Accepted: 03/18/2024] [Indexed: 03/22/2024]
Abstract
Metastatic castration-resistant prostate cancer remains incurable regardless of recent therapeutic advances. Prostate cancer tumors display highly glycolytic phenotypes as the cancer progresses. Nonspecific inhibitors of glycolysis have not been utilized successfully for chemotherapy, because of their penchant to cause systemic toxicity. This study reports the preclinical activity, safety, and pharmacokinetics of a novel small-molecule preclinical candidate, BKIDC-1553, with antiglycolytic activity. We tested a large battery of prostate cancer cell lines for inhibition of cell proliferation, in vitro. Cell-cycle, metabolic, and enzymatic assays were used to demonstrate their mechanism of action. A human patient-derived xenograft model implanted in mice and a human organoid were studied for sensitivity to our BKIDC preclinical candidate. A battery of pharmacokinetic experiments, absorption, distribution, metabolism, and excretion experiments, and in vitro and in vivo toxicology experiments were carried out to assess readiness for clinical trials. We demonstrate a new class of small-molecule inhibitors where antiglycolytic activity in prostate cancer cell lines is mediated through inhibition of hexokinase 2. These compounds display selective growth inhibition across multiple prostate cancer models. We describe a lead BKIDC-1553 that demonstrates promising activity in a preclinical xenograft model of advanced prostate cancer, equivalent to that of enzalutamide. BKIDC-1553 demonstrates safety and pharmacologic properties consistent with a compound that can be taken into human studies with expectations of a good safety margin and predicted dosing for efficacy. This work supports testing BKIDC-1553 and its derivatives in clinical trials for patients with advanced prostate cancer.
Collapse
Affiliation(s)
- Takuma Uo
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - Kayode K. Ojo
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease (CERID), University of Washington; Seattle, Washington 98109, USA
| | - Cynthia C.T. Sprenger
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - Kathryn Soriano Epilepsia
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - B. Gayani K. Perera
- Department of Chemistry, University of Washington; Seattle, Washington 98195, USA
| | - Mamatha Damodarasamy
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - Shihua Sun
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - Soojin Kim
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - Hannah H. Hogan
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - Matthew A. Hulverson
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease (CERID), University of Washington; Seattle, Washington 98109, USA
| | - Ryan Choi
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease (CERID), University of Washington; Seattle, Washington 98109, USA
| | - Grant R. Whitman
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease (CERID), University of Washington; Seattle, Washington 98109, USA
| | - Lynn K. Barrett
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease (CERID), University of Washington; Seattle, Washington 98109, USA
| | - Samantha A. Michaels
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease (CERID), University of Washington; Seattle, Washington 98109, USA
| | - Linda H. Xu
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - Vicky L. Sun
- Department of Pharmaceutics, University of Washington; Seattle, Washington 98195, USA
| | - Samuel L.M. Arnold
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
- Department of Pharmaceutics, University of Washington; Seattle, Washington 98195, USA
| | - Haley J. Pang
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - Matthew M. Nguyen
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
| | - Anna-Lena B.G. Vigil
- Human Biology Division, Fred Hutchinson Cancer Center; Seattle, Washington 98109, USA
| | - Varun Kamat
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, Diabetes Center, University of Washington; Seattle, Washington 98109, USA
| | - Lucas B. Sullivan
- Human Biology Division, Fred Hutchinson Cancer Center; Seattle, Washington 98109, USA
- Department of Biochemistry, University of Washington; Seattle, Washington 98195, USA
| | - Ian R. Sweet
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, Diabetes Center, University of Washington; Seattle, Washington 98109, USA
| | - Ram Vidadala
- Department of Chemistry, University of Washington; Seattle, Washington 98195, USA
| | - Dustin J. Maly
- Department of Chemistry, University of Washington; Seattle, Washington 98195, USA
| | - Wesley C. Van Voorhis
- Department of Medicine, Division of Allergy and Infectious Disease, Center for Emerging and Reemerging Infectious Disease (CERID), University of Washington; Seattle, Washington 98109, USA
| | - Stephen R. Plymate
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington; Seattle, Washington 98109, USA
- Geriatrics Research Education and Clinical Center, VA Puget Sound Health Care System; Seattle, Washington 98108, USA
| |
Collapse
|
6
|
Elbialy A, Kappala D, Desai D, Wang P, Fadiel A, Wang SJ, Makary MS, Lenobel S, Sood A, Gong M, Dason S, Shabsigh A, Clinton S, Parwani AV, Putluri N, Shvets G, Li J, Liu X. Patient-Derived Conditionally Reprogrammed Cells in Prostate Cancer Research. Cells 2024; 13:1005. [PMID: 38920635 PMCID: PMC11201841 DOI: 10.3390/cells13121005] [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: 03/17/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/27/2024] Open
Abstract
Prostate cancer (PCa) remains a leading cause of mortality among American men, with metastatic and recurrent disease posing significant therapeutic challenges due to a limited comprehension of the underlying biological processes governing disease initiation, dormancy, and progression. The conventional use of PCa cell lines has proven inadequate in elucidating the intricate molecular mechanisms driving PCa carcinogenesis, hindering the development of effective treatments. To address this gap, patient-derived primary cell cultures have been developed and play a pivotal role in unraveling the pathophysiological intricacies unique to PCa in each individual, offering valuable insights for translational research. This review explores the applications of the conditional reprogramming (CR) cell culture approach, showcasing its capability to rapidly and effectively cultivate patient-derived normal and tumor cells. The CR strategy facilitates the acquisition of stem cell properties by primary cells, precisely recapitulating the human pathophysiology of PCa. This nuanced understanding enables the identification of novel therapeutics. Specifically, our discussion encompasses the utility of CR cells in elucidating PCa initiation and progression, unraveling the molecular pathogenesis of metastatic PCa, addressing health disparities, and advancing personalized medicine. Coupled with the tumor organoid approach and patient-derived xenografts (PDXs), CR cells present a promising avenue for comprehending cancer biology, exploring new treatment modalities, and advancing precision medicine in the context of PCa. These approaches have been used for two NCI initiatives (PDMR: patient-derived model repositories; HCMI: human cancer models initiatives).
Collapse
Affiliation(s)
- Abdalla Elbialy
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Computational Oncology Unit, The University of Chicago Comprehensive Cancer Center, 900 E 57th Street, KCBD Bldg., STE 4144, Chicago, IL 60637, USA
| | - Deepthi Kappala
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
| | - Dhruv Desai
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
| | - Peng Wang
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
| | - Ahmed Fadiel
- Computational Oncology Unit, The University of Chicago Comprehensive Cancer Center, 900 E 57th Street, KCBD Bldg., STE 4144, Chicago, IL 60637, USA
| | - Shang-Jui Wang
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Mina S. Makary
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Division of Vascular and Interventional Radiology, Department of Radiology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Scott Lenobel
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Division of Musculoskeletal Imaging, Department of Radiology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Akshay Sood
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Department of Urology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Michael Gong
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Department of Urology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Shawn Dason
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Department of Urology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Ahmad Shabsigh
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Department of Urology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Steven Clinton
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
| | - Anil V. Parwani
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Departments of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Gennady Shvets
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14850, USA
| | - Jenny Li
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Departments of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Xuefeng Liu
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Departments of Pathology, Urology, and Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
7
|
Brennen WN, Le Magnen C, Karkampouna S, Anselmino N, Bock N, Choo N, Clark AK, Coleman IM, Dolgos R, Ferguson AM, Goode DL, Krutihof-de Julio M, Navone NM, Nelson PS, O'Neill E, Porter LH, Ranasinghe W, Sunada T, Williams ED, Butler LM, Corey E, van Weerden WM, Taylor RA, Risbridger GP, Lawrence MG. Defining the challenges and opportunities for using patient-derived models in prostate cancer research. Prostate 2024; 84:623-635. [PMID: 38450798 PMCID: PMC11014775 DOI: 10.1002/pros.24682] [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: 12/14/2023] [Revised: 01/29/2024] [Accepted: 02/15/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND There are relatively few widely used models of prostate cancer compared to other common malignancies. This impedes translational prostate cancer research because the range of models does not reflect the diversity of disease seen in clinical practice. In response to this challenge, research laboratories around the world have been developing new patient-derived models of prostate cancer, including xenografts, organoids, and tumor explants. METHODS In May 2023, we held a workshop at the Monash University Prato Campus for researchers with expertise in establishing and using a variety of patient-derived models of prostate cancer. This review summarizes our collective ideas on how patient-derived models are currently being used, the common challenges, and future opportunities for maximizing their usefulness in prostate cancer research. RESULTS An increasing number of patient-derived models for prostate cancer are being developed. Despite their individual limitations and varying success rates, these models are valuable resources for exploring new concepts in prostate cancer biology and for preclinical testing of potential treatments. Here we focus on the need for larger collections of models that represent the changing treatment landscape of prostate cancer, robust readouts for preclinical testing, improved in vitro culture conditions, and integration of the tumor microenvironment. Additional priorities include ensuring model reproducibility, standardization, and replication, and streamlining the exchange of models and data sets among research groups. CONCLUSIONS There are several opportunities to maximize the impact of patient-derived models on prostate cancer research. We must develop large, diverse and accessible cohorts of models and more sophisticated methods for emulating the intricacy of patient tumors. In this way, we can use the samples that are generously donated by patients to advance the outcomes of patients in the future.
Collapse
Affiliation(s)
- W Nathaniel Brennen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, Maryland, USA
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Pharmacology & Molecular Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Clémentine Le Magnen
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Urology, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sofia Karkampouna
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Nicolas Anselmino
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nathalie Bock
- School of Biomedical Sciences at Translational Research Institute, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Max Planck Queensland Centre for the Materials Science of Extracellular Matrices, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
| | - Nicholas Choo
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute Cancer Program, Monash University, Clayton, VIC, Australia
| | - Ashlee K Clark
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute Cancer Program, Monash University, Clayton, VIC, Australia
| | - Ilsa M Coleman
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Robin Dolgos
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Urology, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Alison M Ferguson
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Katharina Gaus Light Microscopy Facility, Mark Wainwright Analytical Centre, Division of Research and Enterprise, University of New South Wales, Sydney, NSW, Australia
| | - David L Goode
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Marianna Krutihof-de Julio
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, Translational Organoid Resource, University of Bern, Bern, Switzerland
| | - Nora M Navone
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Edward O'Neill
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Laura H Porter
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute Cancer Program, Monash University, Clayton, VIC, Australia
| | - Weranja Ranasinghe
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute Cancer Program, Monash University, Clayton, VIC, Australia
- Department of Surgery, Monash University, Melbourne, VIC, Australia
- Department of Urology, Monash Health, Melbourne, VIC, Australia
- Department of Urology, Austin Health, Melbourne, VIC, Australia
| | - Takuro Sunada
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Elizabeth D Williams
- School of Biomedical Sciences at Translational Research Institute, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Australian Prostate Cancer Research Centre-Queensland, Brisbane, QLD, Australia
- Centre for Genomics and Personalised Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Lisa M Butler
- South Australian Immunogenomics Cancer Institute, University of Adelaide, Adelaide, SA, Australia
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington, USA
| | | | - Renea A Taylor
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Physiology, Biomedicine Discovery Institute Cancer Program, Monash University, Clayton, VIC, Australia
- Cabrini Institute, Cabrini Health, Malvern, VIC, Australia
- Melbourne Urological Research Alliance, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Gail P Risbridger
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute Cancer Program, Monash University, Clayton, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Cabrini Institute, Cabrini Health, Malvern, VIC, Australia
- Melbourne Urological Research Alliance, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Mitchell G Lawrence
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute Cancer Program, Monash University, Clayton, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Cabrini Institute, Cabrini Health, Malvern, VIC, Australia
- Melbourne Urological Research Alliance, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
8
|
Wang Y, Xue H, Zhu X, Lin D, Dong X, Chen Z, Chen J, Shi M, Ni Y, Cao J, Wu R, Kang N, Pang X, Crea F, Lin YY, Collins CC, Gleave ME, Parolia A, Chinnaiyan A, Ong CJ, Wang Y. Deciphering the Transcription Factor Landscape in Neuroendocrine Prostate Cancer Progression: A Novel Approach to Understand NE Transdifferentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.27.591428. [PMID: 38746377 PMCID: PMC11092479 DOI: 10.1101/2024.04.27.591428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Background and Objective Prostate cancer (PCa) is a leading cause of cancer mortality in men, with neuroendocrine prostate cancer (NEPC) representing a particularly resistant subtype. The role of transcription factors (TFs) in the progression from prostatic adenocarcinoma (PRAD) to NEPC is poorly understood. This study aims to identify and analyze lineage-specific TF profiles in PRAD and NEPC and illustrate their dynamic shifts during NE transdifferentiation. Methods A novel algorithmic approach was developed to evaluate the weighted expression of TFs within patient samples, enabling a nuanced understanding of TF landscapes in PCa progression and TF dynamic shifts during NE transdifferentiation. Results unveiled TF profiles for PRAD and NEPC, identifying 126 shared TFs, 46 adenocarcinoma-TFs, and 56 NEPC-TFs. Enrichment analysis across multiple clinical cohorts confirmed the lineage specificity and clinical relevance of these lineage-TFs signatures. Functional analysis revealed that lineage-TFs are implicated in pathways critical to cell development, differentiation, and lineage determination. Novel lineage-TF candidates were identified, offering potential targets for therapeutic intervention. Furthermore, our longitudinal study on NE transdifferentiation highlighted dynamic TF expression shifts and delineated a three-phase hypothesis for the process comprised of de-differentiation, dormancy, and re-differentiation. and proposing novel insights into the mechanisms of PCa progression. Conclusion The lineage-specific TF profiles in PRAD and NEPC reveal a dynamic shift in the TF landscape during PCa progression, highlighting three distinct phases of NE transdifferentiation.
Collapse
|
9
|
Liu S, Nam HS, Zeng Z, Deng X, Pashaei E, Zang Y, Yang L, Li C, Huang J, Wendt MK, Lu X, Huang R, Wan J. CDHu40: a novel marker gene set of neuroendocrine prostate cancer (NEPC). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.28.587205. [PMID: 38585861 PMCID: PMC10996696 DOI: 10.1101/2024.03.28.587205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Prostate cancer (PCa) is the most prevalent cancer affecting American men. Castration-resistant prostate cancer (CRPC) can emerge during hormone therapy for PCa, manifesting with elevated serum prostate-specific antigen (PSA) levels, continued disease progression, and/or metastasis to the new sites, resulting in a poor prognosis. A subset of CRPC patients shows a neuroendocrine (NE) phenotype, signifying reduced or no reliance on androgen receptor (AR) signaling and a particularly unfavorable prognosis. In this study, we incorporated computational approaches based on both gene expression profiles and protein-protein interaction (PPI) networks. We identified 500 potential marker genes, which are significantly enriched in cell cycle and neuronal processes. The top 40 candidates, collectively named as CDHu40, demonstrated superior performance in distinguishing NE prostate cancer (NEPC) and non-NEPC samples based on gene expression profiles compared to other published marker sets. Notably, some novel marker genes in CDHu40, absent in the other marker sets, have been reported to be associated with NEPC in the literature, such as DDC, FOLH1, BEX1, MAST1, and CACNA1A. Importantly, elevated CDHu40 scores derived from our predictive model showed a robust correlation with unfavorable survival outcomes in patients, indicating the potential of the CDHu40 score as a promising indicator for predicting the survival prognosis of those patients with the NE phenotype. Motif enrichment analysis on the top candidates suggests that REST and E2F6 may serve as key regulators in the NEPC progression. Significance our study integrates gene expression variances in multiple NEPC studies and protein-protein interaction network to pinpoint a specific set of NEPC maker genes namely CDHu40. These genes and scores based on their gene expression levels effectively distinguish NEPC samples and underscore the clinical prognostic significance and potential mechanism.
Collapse
|
10
|
Labrecque MP, Brown LG, Coleman IM, Nguyen HM, Dalrymple S, Brennen WN, Isaacs JT, Li D, Lakely B, DeLucia DC, Lee JK, Schweizer MT, Lin DW, Corey E, Nelson PS, Morrissey C. Targeting the fibroblast growth factor pathway in molecular subtypes of castration-resistant prostate cancer. Prostate 2024; 84:100-110. [PMID: 37796107 PMCID: PMC10851871 DOI: 10.1002/pros.24630] [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: 06/30/2023] [Revised: 09/06/2023] [Accepted: 09/19/2023] [Indexed: 10/06/2023]
Abstract
BACKGROUND Androgen receptor (AR) pathway inhibition remains the cornerstone for prostate cancer therapies. However, castration-resistant prostate cancer (CRPC) tumors can resist AR signaling inhibitors through AR amplification and AR splice variants in AR-positive CRPC (ARPC), and conversion to AR-null phenotypes, such as double-negative prostate cancer (DNPC) and small cell or neuroendocrine prostate cancer (SCNPC). We have shown previously that DNPC can bypass AR-dependence through fibroblast growth factor receptor (FGFR) signaling. However, the role of the FGFR pathway in other CRPC phenotypes has not been elucidated. METHODS RNA-Seq analysis was conducted on patient metastases, LuCaP patient-derived xenograft (PDX) models, and CRPC cell lines. Cell lines (C4-2B, VCaP, and 22Rv1) and ex vivo LuCaP PDX tumor cells were treated with enzalutamide (ENZA) and FGFR inhibitors (FGFRi) alone or in combination and sensitivity was determined using cell viability assays. In vivo efficacy of FGFRi in ARPC, DNPC, and SCNPC were evaluated using PDX models. RESULTS RNA-Seq analysis of FGFR signaling in metastatic specimens, LuCaP PDX models, and CRPC cell lines revealed significant FGF pathway activation in AR-low PC (ARLPC), DNPC, and SCNPC tumors. In vitro/ex vivo analysis of erdafitinib and CH5183284 demonstrated robust and moderate growth suppression of ARPC, respectively. In vivo studies using four ARPC PDX models showed that combination ENZA and CH5183284 significantly suppressed tumor growth. Additional in vivo studies using four ARPC PDX models revealed that erdafitinib monotherapy was as effective as ENZA in suppressing tumor growth, and there was limited combination benefit. Furthermore, two of three DNPC models and two of four SCNPC models responded to CH5183284 monotherapy, suggesting FGFRi responses were model dependent. RNA-Seq and gene set enrichment analysis of end-of-study ARPC tumors treated with FGFRi displayed decreased expression of E2F and MYC target genes and suppressed G2M checkpoint genes, whereas end-of-study SCNPC tumors had heterogeneous transcriptional responses. CONCLUSIONS Although FGFRi treatments suppressed tumor growth across CRPC phenotypes, our analyses did not identify a single pathway or biomarker that would identify tumor response to FGFRi. This is very likely due to the array of FGFR1-4 expression and tumor phenotypes present in CRPC. Nevertheless, our data nominate the FGFR pathway as a clinically actionable target that promotes tumor growth in diverse phenotypes of treatment-refractory metastatic CRPC.
Collapse
Affiliation(s)
- Mark P. Labrecque
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Lisha G. Brown
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Ilsa M. Coleman
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Holly M. Nguyen
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Susan Dalrymple
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - W. Nathaniel Brennen
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - John T. Isaacs
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Dapei Li
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Bryce Lakely
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Diana C. DeLucia
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - John K. Lee
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Michael T. Schweizer
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Daniel W. Lin
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Eva Corey
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Peter S. Nelson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Colm Morrissey
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
| |
Collapse
|
11
|
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
|
12
|
Verma P, Shukla N, Kumari S, Ansari M, Gautam NK, Patel GK. Cancer stem cell in prostate cancer progression, metastasis and therapy resistance. Biochim Biophys Acta Rev Cancer 2023; 1878:188887. [PMID: 36997008 DOI: 10.1016/j.bbcan.2023.188887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/18/2023] [Accepted: 03/15/2023] [Indexed: 03/31/2023]
Abstract
Prostate cancer (PCa) is the most diagnosed malignancy in the men worldwide. Cancer stem cells (CSCs) are the sub-population of cells present in the tumor which possess unique properties of self-renewal and multilineage differentiation thus thought to be major cause of therapy resistance, disease relapse, and mortality in several malignancies including PCa. CSCs have also been shown positive for the common stem cells markers such as ALDH EZH2, OCT4, SOX2, c-MYC, Nanog etc. Therefore, isolation and characterization of CSCs specific markers which may discriminate CSCs and normal stem cells are critical to selectively eliminate CSCs. Rapid advances in the field offers a theoretical explanation for many of the enduring uncertainties encompassing the etiology and an optimism for the identification of new stem-cell targets, development of reliable and efficient therapies in the future. The emerging reports have also provided unprecedented insights into CSCs plasticity, quiescence, renewal, and therapeutic response. In this review, we discuss the identification of PCa stem cells, their unique properties, stemness-driving pathways, new diagnostics, and therapeutic interventions.
Collapse
|
13
|
Tang DG. Understanding and targeting prostate cancer cell heterogeneity and plasticity. Semin Cancer Biol 2022; 82:68-93. [PMID: 34844845 PMCID: PMC9106849 DOI: 10.1016/j.semcancer.2021.11.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022]
Abstract
Prostate cancer (PCa) is a prevalent malignancy that occurs primarily in old males. Prostate tumors in different patients manifest significant inter-patient heterogeneity with respect to histo-morphological presentations and molecular architecture. An individual patient tumor also harbors genetically distinct clones in which PCa cells display intra-tumor heterogeneity in molecular features and phenotypic marker expression. This inherent PCa cell heterogeneity, e.g., in the expression of androgen receptor (AR), constitutes a barrier to the long-term therapeutic efficacy of AR-targeting therapies. Furthermore, tumor progression as well as therapeutic treatments induce PCa cell plasticity such that AR-positive PCa cells may turn into AR-negative cells and prostate tumors may switch lineage identity from adenocarcinomas to neuroendocrine-like tumors. This induced PCa cell plasticity similarly confers resistance to AR-targeting and other therapies. In this review, I first discuss PCa from the perspective of an abnormal organ development and deregulated cellular differentiation, and discuss the luminal progenitor cells as the likely cells of origin for PCa. I then focus on intrinsic PCa cell heterogeneity in treatment-naïve tumors with the presence of prostate cancer stem cells (PCSCs). I further elaborate on PCa cell plasticity induced by genetic alterations and therapeutic interventions, and present potential strategies to therapeutically tackle PCa cell heterogeneity and plasticity. My discussions will make it clear that, to achieve enduring clinical efficacy, both intrinsic PCa cell heterogeneity and induced PCa cell plasticity need to be targeted with novel combinatorial approaches.
Collapse
Affiliation(s)
- Dean G Tang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Experimental Therapeutics (ET) Graduate Program, The University at Buffalo & Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| |
Collapse
|
14
|
Shi M, Wang Y, Lin D, Wang Y. Patient-derived xenograft models of neuroendocrine prostate cancer. Cancer Lett 2022; 525:160-169. [PMID: 34767925 DOI: 10.1016/j.canlet.2021.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 12/21/2022]
Abstract
In recent years, patient-derived xenografts (PDXs) have attracted much attention as clinically relevant models for basic and translational cancer research. PDXs retain the principal histopathological and molecular heterogeneity of their donor tumors and remain stable across passages. These characteristics allow PDXs to offer a reliable platform for better understanding cancer biology, discovering biomarkers and therapeutic targets, and developing novel therapies. A growing interest in generating neuroendocrine prostate cancer (NEPC) PDX models has been demonstrated, and such models have proven useful in several areas. This review provides a comprehensive summary of currently available NEPC PDX collections, encompassing 1) primary or secondary sites where patient samples were collected, 2) donor patients' treatment histories, 3) morphological features (i.e., small cell and large cell), and 4) genomic alterations. We also highlight suitable models for various research purposes, including identifying therapeutic targets and evaluating drug responses in models with specific genomic backgrounds. Finally, we provide perspectives on the current knowledge gaps and shed light on future applications and improvements of NEPC PDXs.
Collapse
Affiliation(s)
- Mingchen Shi
- Vancouver Prostate Centre, Vancouver, BC, Canada; Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada
| | - Yu Wang
- Vancouver Prostate Centre, Vancouver, BC, Canada; Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada
| | - Dong Lin
- Vancouver Prostate Centre, Vancouver, BC, Canada; Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, BC, Canada; Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada.
| |
Collapse
|
15
|
Wang Y, Wang Y, Ci X, Choi SYC, Crea F, Lin D, Wang Y. Molecular events in neuroendocrine prostate cancer development. Nat Rev Urol 2021; 18:581-596. [PMID: 34290447 PMCID: PMC10802813 DOI: 10.1038/s41585-021-00490-0] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2021] [Indexed: 02/07/2023]
Abstract
Neuroendocrine prostate cancer (NEPC) is a lethal subtype of prostate cancer. NEPC arises de novo only rarely; the disease predominantly develops from adenocarcinoma in response to drug-induced androgen receptor signalling inhibition, although the mechanisms behind this transdifferentiation are a subject of debate. The survival of patients with NEPC is poor, and few effective treatment options are available. To improve clinical outcomes, understanding of the biology and molecular mechanisms regulating NEPC development is crucial. Various NEPC molecular drivers make temporal contributions during NEPC development, and despite the limited treatment options available, several novel targeted therapeutics are currently under research.
Collapse
Affiliation(s)
- Yong Wang
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yu Wang
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada
| | - Xinpei Ci
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada
| | - Stephen Y C Choi
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada
| | - Francesco Crea
- School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Dong Lin
- Vancouver Prostate Centre, Vancouver, BC, Canada.
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada.
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, BC, Canada.
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada.
| |
Collapse
|
16
|
Risbridger GP, Clark AK, Porter LH, Toivanen R, Bakshi A, Lister NL, Pook D, Pezaro CJ, Sandhu S, Keerthikumar S, Quezada Urban R, Papargiris M, Kraska J, Madsen HB, Wang H, Richards MG, Niranjan B, O'Dea S, Teng L, Wheelahan W, Li Z, Choo N, Ouyang JF, Thorne H, Devereux L, Hicks RJ, Sengupta S, Harewood L, Iddawala M, Azad AA, Goad J, Grummet J, Kourambas J, Kwan EM, Moon D, Murphy DG, Pedersen J, Clouston D, Norden S, Ryan A, Furic L, Goode DL, Frydenberg M, Lawrence MG, Taylor RA. The MURAL collection of prostate cancer patient-derived xenografts enables discovery through preclinical models of uro-oncology. Nat Commun 2021; 12:5049. [PMID: 34413304 PMCID: PMC8376965 DOI: 10.1038/s41467-021-25175-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023] Open
Abstract
Preclinical testing is a crucial step in evaluating cancer therapeutics. We aimed to establish a significant resource of patient-derived xenografts (PDXs) of prostate cancer for rapid and systematic evaluation of candidate therapies. The PDX collection comprises 59 tumors collected from 30 patients between 2012-2020, coinciding with availability of abiraterone and enzalutamide. The PDXs represent the clinico-pathological and genomic spectrum of prostate cancer, from treatment-naïve primary tumors to castration-resistant metastases. Inter- and intra-tumor heterogeneity in adenocarcinoma and neuroendocrine phenotypes is evident from bulk and single-cell RNA sequencing data. Organoids can be cultured from PDXs, providing further capabilities for preclinical studies. Using a 1 x 1 x 1 design, we rapidly identify tumors with exceptional responses to combination treatments. To govern the distribution of PDXs, we formed the Melbourne Urological Research Alliance (MURAL). This PDX collection is a substantial resource, expanding the capacity to test and prioritize effective treatments for prospective clinical trials in prostate cancer.
Collapse
Affiliation(s)
- Gail P Risbridger
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia. .,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
| | - Ashlee K Clark
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Laura H Porter
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Roxanne Toivanen
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Andrew Bakshi
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Natalie L Lister
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - David Pook
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC, Australia.,Department of Medical Oncology, Monash Health, Clayton, VIC, Australia
| | - Carmel J Pezaro
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Eastern Health and Monash University Eastern Health Clinical School, Box Hill, VIC, Australia.,Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, England
| | - Shahneen Sandhu
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Cancer Tissue Collection After Death (CASCADE) Program, Melbourne, VIC, Australia
| | - Shivakumar Keerthikumar
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Rosalia Quezada Urban
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Melissa Papargiris
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Australian Prostate Cancer Bioresource, VIC Node, Monash University, Clayton, VIC, Australia
| | - Jenna Kraska
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Australian Prostate Cancer Bioresource, VIC Node, Monash University, Clayton, VIC, Australia
| | - Heather B Madsen
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Australian Prostate Cancer Bioresource, VIC Node, Monash University, Clayton, VIC, Australia
| | - Hong Wang
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Michelle G Richards
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Birunthi Niranjan
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Samantha O'Dea
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Linda Teng
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - William Wheelahan
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Zhuoer Li
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Nicholas Choo
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - John F Ouyang
- Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Heather Thorne
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Lisa Devereux
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Rodney J Hicks
- Center for Molecular Imaging, Peter MacCallum Cancer Center, Melbourne, VIC, Australia
| | - Shomik Sengupta
- Eastern Health and Monash University Eastern Health Clinical School, Box Hill, VIC, Australia.,Department of Urology, Austin Hospital, The University of Melbourne, Heidelberg, VIC, Australia.,Department of Surgery, Austin Health, The University of Melbourne, Heidelberg, VIC, Australia.,Epworth Healthcare, Melbourne, VIC, Australia.,Epworth Freemasons, Epworth Health, East Melbourne, VIC, Australia
| | - Laurence Harewood
- Epworth Healthcare, Melbourne, VIC, Australia.,Department of Surgery, The University of Melbourne, Parkville, VIC, Australia
| | - Mahesh Iddawala
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Arun A Azad
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Jeremy Goad
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Epworth Healthcare, Melbourne, VIC, Australia.,Division of Cancer Surgery, Peter MacCallum Cancer Centre, The University of Melbourne, Melbourne, VIC, Australia
| | - Jeremy Grummet
- Epworth Healthcare, Melbourne, VIC, Australia.,Department of Surgery, Central Clinical School, Monash University, Clayton, VIC, Australia.,Australian Urology Associates, Melbourne, VIC, Australia
| | - John Kourambas
- Department of Medicine, Monash Health, Casey Hospital, Berwick, VIC, Australia
| | - Edmond M Kwan
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC, Australia.,Department of Medical Oncology, Monash Health, Clayton, VIC, Australia
| | - Daniel Moon
- Epworth Healthcare, Melbourne, VIC, Australia.,Division of Cancer Surgery, Peter MacCallum Cancer Centre, The University of Melbourne, Melbourne, VIC, Australia.,Australian Urology Associates, Melbourne, VIC, Australia.,Central Clinical School, Monash University, Clayton, VIC, Australia.,The Epworth Prostate Centre, Epworth Hospital, Richmond, VIC, Australia
| | - Declan G Murphy
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Epworth Healthcare, Melbourne, VIC, Australia.,Division of Cancer Surgery, Peter MacCallum Cancer Centre, The University of Melbourne, Melbourne, VIC, Australia
| | - John Pedersen
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,TissuPath, Mount Waverley, VIC, Australia
| | | | - Sam Norden
- TissuPath, Mount Waverley, VIC, Australia
| | | | - Luc Furic
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - David L Goode
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Mark Frydenberg
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Epworth Healthcare, Melbourne, VIC, Australia.,Australian Urology Associates, Melbourne, VIC, Australia.,Department of Surgery, Monash University, Clayton, VIC, Australia.,Department of Urology, Cabrini Institute, Cabrini Health, Melbourne, VIC, Australia
| | - Mitchell G Lawrence
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Renea A Taylor
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia. .,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia. .,Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Physiology, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
17
|
Devlies W, Handle F, Devos G, Joniau S, Claessens F. Preclinical Models in Prostate Cancer: Resistance to AR Targeting Therapies in Prostate Cancer. Cancers (Basel) 2021; 13:915. [PMID: 33671614 PMCID: PMC7926818 DOI: 10.3390/cancers13040915] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/18/2022] Open
Abstract
Prostate cancer is an androgen-driven tumor. Different prostate cancer therapies consequently focus on blocking the androgen receptor pathway. Clinical studies reported tumor resistance mechanisms by reactivating and bypassing the androgen pathway. Preclinical models allowed the identification, confirmation, and thorough study of these pathways. This review looks into the current and future role of preclinical models to understand resistance to androgen receptor-targeted therapies. Increasing knowledge on this resistance will greatly improve insights into tumor pathophysiology and future treatment strategies in prostate cancer.
Collapse
Affiliation(s)
- Wout Devlies
- Laboratory of Molecular Endocrinology, KU Leuven, 3000 Leuven, Belgium;
- Department of Urology, University Hospitals Leuven, 3000 Leuven, Belgium; (G.D.); (S.J.)
| | - Florian Handle
- Division of Experimental Urology, Department of Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Gaëtan Devos
- Department of Urology, University Hospitals Leuven, 3000 Leuven, Belgium; (G.D.); (S.J.)
| | - Steven Joniau
- Department of Urology, University Hospitals Leuven, 3000 Leuven, Belgium; (G.D.); (S.J.)
| | - Frank Claessens
- Laboratory of Molecular Endocrinology, KU Leuven, 3000 Leuven, Belgium;
| |
Collapse
|
18
|
Kaarijärvi R, Kaljunen H, Ketola K. Molecular and Functional Links between Neurodevelopmental Processes and Treatment-Induced Neuroendocrine Plasticity in Prostate Cancer Progression. Cancers (Basel) 2021; 13:cancers13040692. [PMID: 33572108 PMCID: PMC7915380 DOI: 10.3390/cancers13040692] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Treatment-induced neuroendocrine prostate cancer (t-NEPC) is a subtype of castration-resistant prostate cancer (CRPC) which develops under prolonged androgen deprivation therapy. The mechanisms and pathways underlying the t-NEPC are still poorly understood and there are no effective treatments available. Here, we summarize the literature on the molecules and pathways contributing to neuroendocrine phenotype in prostate cancer in the context of their known cellular neurodevelopmental processes. We also discuss the role of tumor microenvironment in neuroendocrine plasticity, future directions, and therapeutic options under clinical investigation for neuroendocrine prostate cancer. Abstract Neuroendocrine plasticity and treatment-induced neuroendocrine phenotypes have recently been proposed as important resistance mechanisms underlying prostate cancer progression. Treatment-induced neuroendocrine prostate cancer (t-NEPC) is highly aggressive subtype of castration-resistant prostate cancer which develops for one fifth of patients under prolonged androgen deprivation. In recent years, understanding of molecular features and phenotypic changes in neuroendocrine plasticity has been grown. However, there are still fundamental questions to be answered in this emerging research field, for example, why and how do the prostate cancer treatment-resistant cells acquire neuron-like phenotype. The advantages of the phenotypic change and the role of tumor microenvironment in controlling cellular plasticity and in the emergence of treatment-resistant aggressive forms of prostate cancer is mostly unknown. Here, we discuss the molecular and functional links between neurodevelopmental processes and treatment-induced neuroendocrine plasticity in prostate cancer progression and treatment resistance. We provide an overview of the emergence of neurite-like cells in neuroendocrine prostate cancer cells and whether the reported t-NEPC pathways and proteins relate to neurodevelopmental processes like neurogenesis and axonogenesis during the development of treatment resistance. We also discuss emerging novel therapeutic targets modulating neuroendocrine plasticity.
Collapse
|
19
|
Devlies W, Eckstein M, Cimadamore A, Devos G, Moris L, Van den Broeck T, Montironi R, Joniau S, Claessens F, Gevaert T. Clinical Actionability of the Genomic Landscape of Metastatic Castration Resistant Prostate Cancer. Cells 2020; 9:E2494. [PMID: 33212909 PMCID: PMC7698403 DOI: 10.3390/cells9112494] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/02/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022] Open
Abstract
The development of targeted therapies increases treatment options for metastatic castration resistant prostate cancer (mCRPC) patients. There is a need for strong predictive and prognostic signatures to guide physicians in treating mCRPC patients. In this review we unravel the possible actionability in the AR pathway, PI3K AKT signaling, and DNA repair pathways. Additionally, we make recommendations on biomarker trial design, and the clinical use of this new type of data.
Collapse
Affiliation(s)
- Wout Devlies
- Department of Urology, University Hospitals Leuven, 3000 Leuven, Belgium; (G.D.); (L.M.); (T.V.d.B.); (S.J.)
- Laboratory of Molecular Endocrinology, KU Leuven, 3000 Leuven, Belgium;
| | - Markus Eckstein
- Department of Pathology, Friedrich-Alexander-University of Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Alessia Cimadamore
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60121 Ancona, Italy; (A.C.); (R.M.)
| | - Gaëtan Devos
- Department of Urology, University Hospitals Leuven, 3000 Leuven, Belgium; (G.D.); (L.M.); (T.V.d.B.); (S.J.)
| | - Lisa Moris
- Department of Urology, University Hospitals Leuven, 3000 Leuven, Belgium; (G.D.); (L.M.); (T.V.d.B.); (S.J.)
- Laboratory of Molecular Endocrinology, KU Leuven, 3000 Leuven, Belgium;
| | - Thomas Van den Broeck
- Department of Urology, University Hospitals Leuven, 3000 Leuven, Belgium; (G.D.); (L.M.); (T.V.d.B.); (S.J.)
| | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60121 Ancona, Italy; (A.C.); (R.M.)
| | - Steven Joniau
- Department of Urology, University Hospitals Leuven, 3000 Leuven, Belgium; (G.D.); (L.M.); (T.V.d.B.); (S.J.)
| | - Frank Claessens
- Laboratory of Molecular Endocrinology, KU Leuven, 3000 Leuven, Belgium;
| | - Thomas Gevaert
- Department of Pathology, University Hospitals Leuven, 3000 Leuven, Belgium;
| |
Collapse
|
20
|
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
|