Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 Effective treatment development for prostate cancer is hampered by the lack of patient-specific in vitro models that accurately recapitulate this disease. This review focusses on current prostate cancer preclinical models and how recent developmetns and t + 771 word(s) 771 2020-02-06 10:22:23 |
2 format change -1 word(s) 770 2020-02-25 05:20:13 | |
3 format change -4 word(s) 766 2020-10-27 03:43:34 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Hepburn, A.C.; Sims, C.H.C.; Buskin, A.; Heer, R. Preclinical Prostate Cancer Research. Encyclopedia. Available online: https://encyclopedia.pub/entry/296 (accessed on 23 April 2024).
Hepburn AC, Sims CHC, Buskin A, Heer R. Preclinical Prostate Cancer Research. Encyclopedia. Available at: https://encyclopedia.pub/entry/296. Accessed April 23, 2024.
Hepburn, Anastasia C., C. H. Cole Sims, Adriana Buskin, Rakesh Heer. "Preclinical Prostate Cancer Research" Encyclopedia, https://encyclopedia.pub/entry/296 (accessed April 23, 2024).
Hepburn, A.C., Sims, C.H.C., Buskin, A., & Heer, R. (2020, February 14). Preclinical Prostate Cancer Research. In Encyclopedia. https://encyclopedia.pub/entry/296
Hepburn, Anastasia C., et al. "Preclinical Prostate Cancer Research." Encyclopedia. Web. 14 February, 2020.
Preclinical Prostate Cancer Research
Edit

We address the challenges of using primary cultures and patient-derived xenografts to study prostate cancer. We describe emerging approaches using primary prostate epithelial cells and prostate organoids and their genetic manipulation for disease modelling. Furthermore, the use of human prostate-derived induced pluripotent stem cells (iPSCs) is highlighted as a promising complimentary approach. Finally, we discuss the manipulation of iPSCs to generate ‘avatars’ for drug disease testing. Specifically, we describe how a conceptual advance through the creation of living biobanks of "genetically engineered cancers" that contain patient-specific driver mutations hold promise for personalised medicine. 

prostate cancer induced pluripotent stem cells organoids patient-derived xenografts primary culture cell lines preclinical model

1. Introduction

One of the key issues hampering the development of effective treatments for prostate cancer is the lack of suitable, tractable, and patient-specific in vitro models that accurately recapitulate this disease.

2. Emerging Approaches

2.1. Transformation of Primary Prostate Cells

In a seminal paper, it was shown that primary benign human basal prostate epithelium can initiate prostate cancer in immune-deficient mice and that the derived tumours realistically recreate histology of in situ human prostate cancer[1]. This has been the basis for an interesting strategy to overcome some of the problems of primary prostate cancer culture, where researchers can now transform easier to grow benign prostate epithelium to generate prostate cancer organoids[2]. These studies lay the platform for a new paradigm, where benign cells are converted into “designer” cancers harbouring specific mutations of interest. These can be repeatedly generated whilst faithfully maintaining the genotypes of interest avoiding the ever evolving subclonal progressions affecting long term culture of primary derived cancers[3]. Despite the promise of this “tumour engineering” approach there are limitations to maintaining even benign prostate epithelial cultures and a more ready supply of cells to manipulate are ideally required. In this respect, the emergence of easy to expand and immortalised iPSCs and the ability to differentiate these in the tissue type of interest offers a new way forward.

2.2. Prostate iDOs

Recently, a high throughput model of generating human prostate organoids from iPSCs has also been described, involving co-culturing iPSCs with rodent urogenital sinus mesenchyme (UGM). This simple differentiation protocol results in glandular structures in vitro that faithfully mimic prostate tissue histology and express key prostate markers such as AR, prostate specific homeobox protein NKX3.1 and secretory prostate specific antigen (PSA)[4]. This approach built on previous data showing the generation of prostate tissue in xenograft studies from ESCs[5]. Differentiation from iPSCs avoids many ethical and regulatory restrictions relating to ESCs and enable greater access to organoid generation to groups worldwide culture[6][7]. Previous in vitro human prostate organoid approaches, from either tissue-derived cells or ESCs, do not recreate the full breadth of in situ prostate differentiation as they do not contain neuroendocrine cells particularly relevant in light of emerging data showing that neuroendocrine differentiation drives treatment-resistant prostate cancer[8][9][10]. Additionally, it would be of interest to determine whether following maturation of prostate iDOs there is a switch to a somatic stem cell mode of homeostasis, identified by the presence of DLK1-enriched basal stem cells, to sustain long-term culture[11]. High-throughput iPSC-derived human prostate tissue generation provides unparalleled scope for in vitro disease modelling and drug discovery without the constraints of tissue accessibility and long-standing difficulties associated with primary culture.

2.3. Genome Editing Technology and Precision Medicine

Genome editing technology has emerged as an extremely powerful tool that can greatly advance organoid-based research for the development of better targeted therapies[12]. CRISPR-Cas9 genome editing induces double-stranded DNA breaks at specific loci adjacent to a protospacer-adjacent motif (PAM) using a complementary single-guide RNA sequence (sgRNA) and Cas9 endonuclease[13]. DNA repair then takes place by either non-homologous end joining, resulting in insertions/deletions (INDELs) or homology-directed repair with a donor template. In 2013, Schwank et al. reported the first successful therapeutic CRISPR-Cas9 gene editing in human tissue, by correcting the CFTR locus in intestinal organoids from patients with cystic fibrosis (CF), making CF treatment a possible reality[14]. Since then, CRISPR-Cas9 has further been used to reproduce genetic mutations that occur in cancers including prostate cancer. In 2017, for the first time, the use of CRISPR/Cas9 to create endogenous gene fusions in organoids was reported[15]. Mouse prostate organoids were modified to carry the TMPRSS2-ERG fusion, a genetic alteration present in more than 50% of prostate cancers that leads to high ERG expression driven from the androgen-responsive promoter of the TMPRSS2 gene[15][16][17]. Previously this fusion had been modelled by artificial ERG overexpression and studied in human prostate cancer cell lines and mouse models, but this approach for the first time allows investigation of its effect in a wildtype background[15][18].

References

  1. Andrew S. Goldstein; Jiaoti Huang; Changyong Guo; Isla P. Garraway; Owen N. Witte; Identification of a Cell of Origin for Human Prostate Cancer. Science 2010, 329, 568-571, 10.1126/science.1189992.
  2. Kenji Unno; Meejeon Roh; Young A. Yoo; Yousef Al-Shraideh; Lu Wang; Larisa Nonn; Sarki A. Abdulkadir; Modeling African American prostate adenocarcinoma by inducing defined genetic alterations in organoids. Oncotarget 2017, 8, 51264-51276, 10.18632/oncotarget.17230.
  3. Sophie F. Roerink; Nobuo Sasaki; Henry Lee-Six; Matthew D. Young; Ludmil B. Alexandrov; Sam Behjati; Thomas J. Mitchell; Sebastian Grossmann; Howard Lightfoot; David A. Egan; et al.Apollo PronkNiels SmakmanJoost Van GorpElizabeth AndersonStephen GambleChris AlderMarc Van De WeteringPeter J. CampbellMichael R. StrattonHans Clevers Intra-tumour diversification in colorectal cancer at the single-cell level. Nature 2018, 556, 457-462, 10.1038/s41586-018-0024-3.
  4. Ac Hepburn; El Curry; Mohammad Moad; Re Steele; Oe Franco; Laura Wilson; Parmveer Singh; Se Crawford; Luke Gaughan; Ig Mills; et al.Sw HaywardCn RobsonRakesh HeerAnastasia HepburnEmma CurryRebecca SteeleOmar FrancoSusan CrawfordIan MillsSimon HaywardCraig Robson High-throughput propagation of human prostate tissue from induced-pluripotent stem cells. bioRxiv 2019, null, 637876, 10.1101/637876.
  5. Renea A. Taylor; Prue A Cowin; Gerald R Cunha; Martin F. Pera; Alan Trounson; John Pedersen; Gail P. Risbridger; Formation of human prostate tissue from embryonic stem cells. Nature Methods 2006, 3, 179-181, 10.1038/nmeth855.
  6. John A Robertson; Human embryonic stem cell research: ethical and legal issues. Nature Reviews Microbiology 2001, 2, 74-78, 10.1038/35047594.
  7. Tomer Halevy; Achia Urbach; Comparing ESC and iPSC—Based Models for Human Genetic Disorders. Journal of Clinical Medicine 2014, 3, 1146-1162, 10.3390/jcm3041146.
  8. Wouter R. Karthaus; Phillip J. Iaquinta; Jarno Drost; Ana Gracanin; Ruben Van Boxtel; John Wongvipat; Catherine M. Dowling; Dong Gao; Harry Begthel; Norman Sachs; et al.Robert G.J. VriesEdwin CuppenYu ChenCharles L. SawyersHans Clevers Identification of multipotent luminal progenitor cells in human prostate organoid cultures.. Cell 2014, 159, 163-175, 10.1016/j.cell.2014.08.017.
  9. Jennifer Bishop; Daksh Thaper; Sepideh Vahid; Alastair Davies; Kirsi Ketola; Hidetoshi Kuruma; Randy Jama; Ka Mun Nip; Arkhjamil Angeles; Fraser Johnson; Alexander W. Wyatt; Ladan Fazli; Martin Gleave4; Ng Lin; Mark A. Rubin; Colin C Collins; Yuzhuo Wang4; Himisha Beltran; Amina Zoubeidi; The Master Neural Transcription Factor BRN2 Is an Androgen Receptor–Suppressed Driver of Neuroendocrine Differentiation in Prostate Cancer. Cancer Discovery 2016, 7, 54-71, 10.1158/2159-8290.CD-15-1263.
  10. Esther L. Calderon-Gierszal; Gail Prins; Directed Differentiation of Human Embryonic Stem Cells into Prostate Organoids In Vitro and its Perturbation by Low-Dose Bisphenol A Exposure. PLOS ONE 2015, 10, e0133238, 10.1371/journal.pone.0133238.
  11. Mohammad Moad; Edouard Hannezo; Simon J.A. Buczacki; Laura Wilson; Amira El-Sherif; David Sims; Robert Pickard; Nicholas A. Wright; Stuart Williamson; Doug M. Turnbull; et al.Robert W. TaylorLaura C. GreavesCraig N. RobsonB. D. SimonsRakesh Heer Multipotent Basal Stem Cells, Maintained in Localized Proximal Niches, Support Directed Long-Ranging Epithelial Flows in Human Prostates.. Cell Reports 2017, 20, 1609-1622, 10.1016/j.celrep.2017.07.061.
  12. Else Driehuis; Hans Clevers; CRISPR/Cas 9 genome editing and its applications in organoids. American Journal of Physiology-Gastrointestinal and Liver Physiology 2017, 312, G257-G265, 10.1152/ajpgi.00410.2016.
  13. L. Cong; F. Ann Ran; David Cox; Shuailiang Lin; Robert Barretto; Naomi Habib; Patrick Hsu; Xuebing Wu; Wenyan Jiang; Luciano A. Marraffini; et al.Feng Zhang Multiplex Genome Engineering Using CRISPR/Cas Systems. Science 2013, 339, 819-823, 10.1126/science.1231143.
  14. Gerald Schwank; Bon-Kyoung Koo; Valentina Sasselli; Johanna F. Dekkers; Inha Heo; Turan Demircan; Nobuo Sasaki; Sander Boymans; Edwin Cuppen; Cornelis K. Van Der Ent; et al.Edward E.S. NieuwenhuisJeffrey M. BeekmanHans Clevers Functional Repair of CFTR by CRISPR/Cas9 in Intestinal Stem Cell Organoids of Cystic Fibrosis Patients. Cell Stem Cell 2013, 13, 653-658, 10.1016/j.stem.2013.11.002.
  15. Else Driehuis; Hans Clevers; CRISPR-Induced TMPRSS2-ERG Gene Fusions in Mouse Prostate Organoids. JSM biotechnology & biomedical engineering 2017, 4, 1076.
  16. F Demichelis; Katja Fall; S Perner; O Andrén; F Schmidt; S R Setlur; Y Hoshida; J-M Mosquera; Y Pawitan; C Lee; H-O Adami; L A Mucci; Philip Kantoff; S-O Andersson; A M Chinnaiyan; J-E Johansson; Mark A. Rubin; Erratum: TMPRSS2:ERG gene fusion associated with lethal prostate cancer in a watchful waiting cohort. Oncogene 2007, 26, 5692-5692, 10.1038/sj.onc.1210630.
  17. S.A. Tomlins; D.R. Rhodes; S. Perner; Saravana Mohan Dhanasekaran; R. Mehra; X.-W. Sun; S. Varambally; X. Cao; J. Tchinda; R. Kuefer; et al.C. LeeJ.E. MontieR.B. ShahK.J. PientaM.A. RubinA.M. Chinnaiyan Recurrent Fusion of TMPRSS2 and ETS Transcription Factor Genes in Prostate Cancer. Science 2005, 310, 644–648, 10.1016/s0022-5347(06)00096-6.
  18. Scott Tomlins; Bharathi Laxman; Sooryanarayana Varambally; Xuhong Cao; Jindan Yu; Beth E. Helgeson; Qi Cao; John Prensner; Mark A. Rubin; Rajal B. Shah; et al.Rohit MehraArul M. Chinnaiyan Role of the TMPRSS2-ERG Gene Fusion in Prostate Cancer. Neoplasia 2008, 10, 177-IN9, 10.1593/neo.07822.
More
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 753
Revisions: 3 times (View History)
Update Date: 27 Oct 2020
1000/1000