Journal Home > Volume 2 , Issue 3

Androgen receptor (AR) signaling have been frequently targeted for treating prostate cancer (PCa). Even though primarily patients receive a good therapeutic outcome by targeting AR signaling axis, eventually it emerges resistance by altering the genetic makeup of prostate cells. However, to develop an effective therapeutic regime, it is essential to recognize key genetic alterations in PCa. The most common genetic alterations that give rise to distinct androgen different differentiation states are gene fusion of TMPRSS2 with ETS family genes, deletion, or mutation of tumor suppressor PTEN and TP53 gene, amplification or splicing of AR, altered DNA repair genes. In this review, we describe key genes and genetic changes that have been recognized to contribute to altered prostate environment.


menu
Abstract
Full text
Outline
About this article

Symphony in the crowd: Key genetic alterations in prostate cancer

Show Author's information Neshat Masud ( )
Department of Pharmacology, University of Louisiana at Monroe, Monroe, LA, USA

Abstract

Androgen receptor (AR) signaling have been frequently targeted for treating prostate cancer (PCa). Even though primarily patients receive a good therapeutic outcome by targeting AR signaling axis, eventually it emerges resistance by altering the genetic makeup of prostate cells. However, to develop an effective therapeutic regime, it is essential to recognize key genetic alterations in PCa. The most common genetic alterations that give rise to distinct androgen different differentiation states are gene fusion of TMPRSS2 with ETS family genes, deletion, or mutation of tumor suppressor PTEN and TP53 gene, amplification or splicing of AR, altered DNA repair genes. In this review, we describe key genes and genetic changes that have been recognized to contribute to altered prostate environment.

Keywords: cancer, prostate cancer, genetic, alterations, symphony

References(39)

1

Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7–33. https://doi.org/10.3322/caac.21708

2

Barilla S, Lindblom A, Helgadottir HT. Unravelling genetic variants of a Swedish family with high risk of prostate cancer. Hereditary Cancer Clin Pract. 2022;20(1):28. https://doi.org/10.1186/s13053-022-00234-0

3

Thompson J, Hyytinen ER, Haapala K, Rantala I, Helin HJ, Jänne OA, et al. Androgen receptor mutations in high‐grade prostate cancer before hormonal therapy. Lab Invest. 2003;83(12):1709–13. https://doi.org/10.1097/01.LAB.0000107262.40402.44

4

Pavel AG, Stambouli D, Gener I, Preda A, Anton G, Baston C. Genetic variant located on chromosome 17p12 contributes to prostate cancer onset and biochemical recurrence. Sci Rep. 2022;12(1):4546. https://doi.org/10.1038/s41598-022-08472-x

5

Eeles RA, Kote‐Jarai Z, Giles GG, Olama AAA, Guy M, Jugurnauth SK, et al. Multiple newly identified loci associated with prostate cancer susceptibility. Nat Genet. 2008;40(3):316–21. https://doi.org/10.1038/ng.90

6

Finch A, Clark R, Vesprini D, Lorentz J, Kim RH, Thain E, et al. An appraisal of genetic testing for prostate cancer susceptibility. NPJ Precision Oncology. 2022;6(1):43. https://doi.org/10.1038/s41698-022-00282-8

7

Sipeky C, Tammela TLJ, Auvinen A, Schleutker J. Novel prostate cancer susceptibility gene SP6 predisposes patients to aggressive disease. Prostate Cancer Prostatic Dis. 2021;24(4):1158–66. https://doi.org/10.1038/s41391-021-00378-5

8

McNeal JE. The zonal anatomy of the prostate. Prostate. 1981;2(1):35–49. https://doi.org/10.1002/pros.2990020105

9

Timms BG. Prostate development: a historical perspective. Differentiation. 2008;76(6):565–77. https://doi.org/10.1111/j.1432-0436.2008.00278.x

10

Rebello RJ, Oing C, Knudsen KE, Loeb S, Johnson DC, Reiter RE, et al. Prostate cancer. Nat Rev Dis Primers. 2021;7:9. https://doi.org/10.1038/s41572-020-00243-0

11

Zhou Q, Nie R, Prins GS, Saunders PT, Katzenellenbogen BS, Hess RA. Localization of androgen and estrogen receptors in adult Male mouse reproductive tract. J Androl. 2002;23(6):870–81. https://doi.org/10.1016/0012-1606(88)90260-6

12

Donjacour AA, Cunha GR. The effect of androgen deprivation on branching morphogenesis in the mouse prostate. Dev Biol. 1988;128(1):1–14. https://doi.org/10.1016/0012-1606(88)90260-6

13

Shen MM, Abate‐Shen C. Molecular genetics of prostate cancer: new prospects for old challenges. Genes Dev. 2010;24(18):1967–2000. https://doi.org/10.1101/gad.1965810

14

Wang L, Dehm SM, Hillman DW, Sicotte H, Tan W, Gormley M, et al. A prospective genome‐wide study of prostate cancer metastases reveals association of WNT pathway activation and increased cell cycle proliferation with primary resistance to abiraterone acetate‐prednisone. Ann Oncol. 2018;29(2):352–60. https://doi.org/10.1093/annonc/mdx689

15

Sakr WA, Grignon DJ, Crissman JD, Heilbrun LK, Cassin BJ, Pontes JJ, et al. High grade prostatic intraepithelial neoplasia (HGPIN) and prostatic adenocarcinoma between the ages of 20‐69: an autopsy study of 249 cases. In Vivo. 1994;8(3):439–43. PMID: 7803731. https://doi.org/10.1038/ng.3419

16

Pomerantz MM, Li F, Takeda DY, Lenci R, Chonkar A, Chabot M, et al. The androgen receptor cistrome is extensively reprogrammed in human prostate tumorigenesis. Nat Genet. 2015;47(11):1346–51. https://doi.org/10.1038/ng.3419

17

Lin C, Yang L, Tanasa B, Hutt K, Ju B, Ohgi KA, et al. Nuclear receptor‐induced chromosomal proximity and DNA breaks underlie specific translocations in cancer. Cell. 2009;139(6):1069–83. https://doi.org/10.1016/j.cell.2009.11.030

18

Haffner MC, De Marzo AM, Meeker AK, Nelson WG, Yegnasubramanian S. Transcription‐induced DNA double strand breaks: both oncogenic force and potential therapeutic target. Clin Cancer Res. 2011;17(12):3858–64. https://doi.org/10.1158/1078-0432.CCR-10-2044

19

Heo SH, Choi YJ, Ryoo HM, Cho JY. Expression profiling of ETS and MMP factors in VEGF‐activated endothelial cells: role of MMP‐10 in VEGF‐induced angiogenesis. J Cell Physiol. 2010;224(3):734–42. https://doi.org/10.1002/jcp.22175

20

Kumar‐Sinha C, Tomlins SA, Chinnaiyan AM. Recurrent gene fusions in prostate cancer. Nat Rev Cancer. 2008;8(7):497–511. https://doi.org/10.1038/nrc2402

21

Tomlins SA, Rhodes DR, Perner S, Dhanasekaran SM, Mehra R, Sun XW, et al. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science. 2005;310(5748):644–8. https://doi.org/10.1126/science.1117679

22

Carver BS, Tran J, Chen Z, Carracedo‐Perez A, Alimonti A, Nardella C, et al. ETS rearrangements and prostate cancer initiation. Nature. 2009;457(7231):E1–E3. https://doi.org/10.1038/nature07738

23

Attard G, Clark J, Ambroisine L, Fisher G, Kovacs G, Flohr P, et al. Duplication of the fusion of TMPRSS2 to ERG sequences identifies fatal human prostate cancer. Oncogene. 2008;27(3):253–63. https://doi.org/10.1038/sj.onc.1210640

24

Krohn A, Diedler T, Burkhardt L, Mayer PS, De Silva C, Meyer‐Kornblum M, et al. Genomic deletion of PTEN is associated with tumor progression and early PSA recurrence in ERG fusion‐positive and fusion‐negative prostate cancer. Am J Pathol. 2012;181(2):401–12. https://doi.org/10.1016/j.ajpath.2012.04.026

25

Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang SI, et al. PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science. 1997;275(5308):1943–7. https://doi.org/10.1126/science.275.5308.1943

26

Wang S, Gao J, Lei Q, Rozengurt N, Pritchard C, Jiao J, et al. Prostate‐specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer. Cancer Cell. 2003;4(3):209–21. https://doi.org/10.1016/s1535-6108(03)00215-0

27

Chen Y, Chi P, Rockowitz S, Iaquinta PJ, Shamu T, Shukla S, et al. ETS factors reprogram the androgen receptor cistrome and prime prostate tumorigenesis in response to PTEN loss. Nat Med. 2013;19(8):1023–9. https://doi.org/10.1038/nm.3216

28

Salvesan HB, MacDonad N, Ryan A, Jacobs IJ, Lynch ED, Akslen AK, et al. PTEN methylation is associated with tumor state and microsatellite instability in endometrial carcinoma. Int J Cancer. 2001;91(1):22–6. https://doi.org/10.1002/1097-0215(20010101)91:1<22::AID-IJC1002>3.0.CO;2-S

29

Cunha GR. The role of androgens in the epithelio‐mesenchymal interactions involved in prostatic morphogenesis in embryonic mice. Anat Rec. 1973;175(1):87–96. https://doi.org/10.1002/ar.1091750108

30

Kallio HML, Hieta R, Latonen L, Brofeldt A, Annala M, Kivinummi K, et al. Constitutively active androgen receptor splice variants AR‐V3, AR‐V7 and AR‐V9 are co‐expressed in castration‐resistant prostate cancer metastases. Br J Cancer. 2018;119(3):347–56. https://doi.org/10.1038/s41416-018-0172-0

31

Hu R, Dunn TA, Wei S, Isharwal S, Veltri RW, Humphreys E, et al. Ligand‐independent androgen receptor variants derived from splicing of cryptic exons signify hormone‐refractory prostate cancer. Cancer Res. 2009;69(1):16–22. https://doi.org/10.1158/0008-5472.CAN-08-2764

32

Barbieri CE, Chinnaiyan AM, Lerner SP, Swanton C, Rubin MA. The emergence of precision urologic oncology: a collaborative review on biomarker‐driven therapeutics. Eur Urol. 2017;71(2):237–46. https://doi.org/10.1016/j.eururo.2016.08.024

33

Sumanasuriya S, De Bono J. Treatment of advanced prostate cancer‐a review of current therapies and future promise. Cold Spring Harbor Perspect Med. 2018;8(6):a030635. https://doi.org/10.1101/cshperspect.a030635

34

Zhao D, Lu X, Wang G, Lan Z, Liao W, Li J, et al. Synthetic essentiality of chromatin remodelling factor CHD1 in PTEN‐deficient cancer. Nature. 2017;542(7642):484–8. https://doi.org/10.1038/nature21357.

35

Shenoy TR, Boysen G, Wang MY, Xu QZ, Guo W, Koh FM, et al. CHD1 loss sensitizes prostate cancer to DNA damaging therapy by promoting error‐prone double‐strand break repair. Ann Oncol. 2017;28(7):1495–507. https://doi.org/10.1093/annonc/mdx165

36

Miyamoto DT, Lee RJ, Stott SL, Ting DT, Wittner BS, Ulman M, et al. Androgen receptor signaling in circulating tumor cells as a marker of hormonally responsive prostate cancer. Cancer Discov. 2012;2(11):995–1003. https://doi.org/10.1158/2159-8290.CD-12-0222

37

Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, et al. Tissue‐based map of the human proteome. Science. 2015;347(6220):1260419. https://doi.org/10.1126/science.1260419

38

Beltran H, Prandi D, Mosquera JM, Benelli M, Puca L, Cyrta J, et al. Divergent clonal evolution of castration resistant neuroendocrine prostate cancer. Nat Med. 2016;22(3):298–305. https://doi.org/10.1038/nm.4045

39

Yousef GM, Obiezu CV, Luo LY, Black MH, Diamandis EP. Prostase/KLK‐L1 is a new member of the human kallikrein gene family, is expressed in prostate and breast tissues, and is hormonally regulated. Cancer Res. 1999;59(17):4252–56. https://doi.org/10.1006/geno.2000.6346

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 11 September 2022
Accepted: 12 January 2023
Published: 09 February 2023
Issue date: June 2023

Copyright

© 2023 The Authors. Tsinghua University Press.

Acknowledgements

ACKNOWLEDGMENTS

None.

Rights and permissions

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

Return