Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-hfldf Total loading time: 0 Render date: 2024-06-08T11:16:32.297Z Has data issue: false hasContentIssue false

52 - Prostate cancer

from Part 3.1 - Molecular pathology: carcinomas

Published online by Cambridge University Press:  05 February 2015

Kaustubh Datta
Affiliation:
Department of Urologic Research, Biochemistry and Molecular Biology, Mayo Clinic Cancer Center, Mayo Clinic Foundation, Rochester, MN, USA
Donald J. Tindall
Affiliation:
Department of Urologic Research, Biochemistry and Molecular Biology, Mayo Clinic Cancer Center, Mayo Clinic Foundation, Rochester, MN, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Introduction

Prostate cancer (PCa) is the most frequently diagnosed, and the second leading cause of death from cancer in North American and European men (1). It has the highest incidence rate among all epithelial tumors in the United States. Approximately 1 in 6 men over the age of 65 is diagnosed with PCa (2). Age, racial background, dietary factors, life-style related factors, and androgens are known contributors to the risk of PCa.

Heterogeneity in PCa

There is a high variability in the natural history of PCa ranging from indolent and asymptomatic to aggressive and metastatic disease. It is also multi-focal and heterogeneous in clinical, histological, and biological dispositions. Healthy glands, precursor, and neoplastic lesions with varying degrees of differentiation often exist in juxtaposition and evolve independently in the same PCa tissue (3,4).

Prostate intra-epithelial neoplasia (PIN) is thought to be a precursor lesion for PCa. High-grade PIN with increased nuclear enlargement, prominent nucleoli, chromatin alterations, and luminal complexity in acinar epithelium is often detected together with PCa in biopsy samples (4,5). PIN shares many common genetic, molecular, and phenotypic characteristics of prostate adenocarcinoma (3,5).

Gleason grade has been used to indicate the severity of the disease.AhigherGleason grade suggests a poorly diferentiated, advanced carcinoma and is used as a prognostic indicator (4).

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 591 - 597
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Deutsch, E, Maggiorella, L, Eschwege, P, et al. Environmental, genetic, and molecular features of prostate cancer. Lancet Oncology 2004;5:303–13.CrossRef
Walsh, PC, DeWeese, TL, Eisenberger, MA.Clinical practice. Localized prostate cancer. New England Journal of Medicine 2007;357:2696–705.CrossRefGoogle ScholarPubMed
Liska, J, Repiska, V, Galbavy, S, et al. Prostate tumours–histological classification and molecular aspects of prostate tumorigenesis. Endocrine Regulations 2007;41:45–57.
Abate-Shen, C, Shen, MM. Molecular genetics of prostate cancer. Genes and Development 2000;14:2410–34.CrossRef
Ayala, AG, Ro, JY. Prostatic intraepithelial neoplasia: recent advances. Archives of Pathology and Laboratory Medicine 2007;131:1257–66.
Morris, DS, Tomlins, SA, Rhodes, DR, et al. Integrating biomedical knowledge to model pathways of prostate cancer progression. Cell Cycle 2007;6:1177–87.CrossRef
MacGrogan, D, Levy, A, Bostwick, D, et al. Loss of chromosome arm 8p loci in prostate cancer: mapping by quantitative allelic imbalance. Genes, Chromosomes and Cancer 1994;10:151–9.CrossRef
Dong, JT. Prevalent mutations in prostate cancer. Journal of Cellular Biochemistry 2006;97:433–47.CrossRefGoogle ScholarPubMed
Nelson, WG, De Marzo, AM, Isaacs, WB.Prostate cancer. New England Journal of Medicine 2003;349:366–81.CrossRefGoogle ScholarPubMed
Lee, WH, Morton, RA, Epstein, JI, et al. Cytidine methylation of regulatory sequences near the pi-class glutathione S-transferase gene accompanies human prostatic carcinogenesis. Proceedings of the National Academy of Sciences USA 1994;91:11 733–7.
Hughes, C, Murphy, A, Martin, C, Sheils, O, O’Leary, J.Molecular pathology of prostate cancer. Journal of Clinical Pathology 2005;58:673–84.CrossRefGoogle ScholarPubMed
Prescott, JL, Blok, L, Tindall, DJ. Isolation and androgen regulation of the human homeobox cDNA, NKX3.1. Prostate 1998;35:71–80.
Bhatia-Gaur, R, Donjacour, AA, Sciavolino, PJ, et al. Roles for Nkx3.1 in prostate development and cancer. Genes and Development 1999;13:966–77.CrossRef
Asatiani, E, Huang, WX, Wang, A, et al. Deletion, methylation, and expression of the NKX3.1 suppressor gene in primary human prostate cancer. Cancer Research 2005;65:1164–73.CrossRef
Bowen, C, Bubendorf, L, Voeller, HJ, et al. Loss of NKX3.1 expression in human prostate cancers correlates with tumor progression. Cancer Research 2000;60:6111–15.
Aslan, G, Irer, B, Tuna, B, et al. Analysis of NKX3.1 expression in prostate cancer tissues and correlation with clinicopathologic features. Pathology Research and Practice 2006;202:93–8.CrossRef
Bethel, CR, Faith, D, Li, X, et al. Decreased NKX3.1 protein expression in focal prostatic atrophy, prostatic intraepithelial neoplasia, and adenocarcinoma: association with gleason score and chromosome 8p deletion. Cancer Research 2006;66:10 683–90.
Morris, DS, Tomlins, SA, Montie, JE, Chinnaiyan, AM.The discovery and application of gene fusions in prostate cancer. British Journal of Urology International 2008;102:276–82.CrossRefGoogle ScholarPubMed
Tomlins, SA, Rhodes, DR, Perner, S, et al. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 2005;310:644–8.CrossRef
Klezovitch, O, Risk, M, Coleman, I, et al. A causal role for ERG in neoplastic transformation of prostate epithelium. Proceedings of the National Academy of Sciences USA 2008;105:2105–10.CrossRef
Clark, JP, Cooper, CS. ETS gene fusions in prostate cancer. Nature Reviews 2009;6:429–39.
Tomlins, SA, Laxman, B, Dhanasekaran, SM, et al. Distinct classes of chromosomal rearrangements create oncogenic ETS gene fusions in prostate cancer. Nature 2007;448:595–9.CrossRef
Carver, BS, Tran, J, Gopalan, A, et al. Aberrant ERG expression cooperates with loss of PTEN to promote cancer progression in the prostate. Nature Genetics 2009;41:619–24.CrossRef
King, JC, Xu, J, Wongvipat, J, et al. Cooperativity of TMPRSS2-ERG with PI3-kinase pathway activation in prostate oncogenesis. Nature Genetics 2009;41:524–6.CrossRef
Dehm, SM, Tindall, DJ. Regulation of androgen receptor signaling in prostate cancer. Expert Review of Anticancer Therapy 2005;5:63–74.CrossRef
Dehm, SM, Tindall, DJ. Androgen receptor structural and functional elements: role and regulation in prostate cancer. Molecular Endocrinology 2007;21:2855–63.CrossRef
Heinlein, CA, Chang, C. Androgen receptor in prostate cancer. Endocrine Reviews 2004;25:276–308.CrossRef
Jain, RK, Safabakhsh, N, Sckell, A, et al. Endothelial cell death, angiogenesis, and microvascular function after castration in an androgen-dependent tumor: role of vascular endothelial growth factor. Proceedings of the National Academy of Sciences USA 1998;95:10 820–5.
Dehm, SM, Tindall, DJ.Molecular regulation of androgen action in prostate cancer. Journal of Cell Biochemistry 2006;99:333–44.CrossRefGoogle ScholarPubMed
Vander Griend, DJ, Litvinov, IV, Isaacs, JT. Stabilizing androgen receptor in mitosis inhibits prostate cancer proliferation. Cell Cycle 2007;6:647–51.CrossRef
Balk, SP, Knudsen, KE. AR, the cell cycle, and prostate cancer. Nuclear Receptor Signaling 2008;6:e001.
Wang, Q, Carroll, JS, Brown, M. Spatial and temporal recruitment of androgen receptor and its coactivators involves chromosomal looping and polymerase tracking. Molecular Cell 2005;19:631–42.CrossRef
Litvinov, IV, Vander Griend, DJ, Antony, L, et al. Androgen receptor as a licensing factor for DNA replication in androgen-sensitive prostate cancer cells. Proceedings of the National Academy of Sciences USA 2006;103:15 085–90.
Mabjeesh, NJ, Willard, MT, Frederickson, CE, Zhong, H, Simons, JW. Androgens stimulate hypoxia-inducible factor 1 activation via autocrine loop of tyrosine kinase receptor/phosphatidylinositol 3’-kinase/protein kinase B in prostate cancer cells. Clinical Cancer Research 2003;9:2416–25.
Rubin, MA. Targeted therapy of cancer: new roles for pathologists–prostate cancer. Modern Pathology 2008;21:S44–55.
Shin, S, Kim, TD, Jin, F, et al. Induction of prostatic intraepithelial neoplasia and modulation of androgen receptor by ETS variant 1/ETS-related protein 81. Cancer Research 2009;69:8102–10.CrossRef
Ribas, J, Ni, X, Haffner, M, et al. miR-21: an androgen receptor-regulated microRNA that promotes hormone-dependent and hormone-independent prostate cancer growth. Cancer Research 2009;69:7165–9.CrossRef
Kaarbo, M, Klokk, TI, Saatcioglu, F. Androgen signaling and its interactions with other signaling pathways in prostate cancer. Bioessays 2007;29:1227–38.CrossRef
Culig, Z, Hobisch, A, Hittmair, A, et al. Androgen receptor gene mutations in prostate cancer: implications for disease progression and therapy. Drugs and Aging 1997;10:50–8.CrossRef
Mohler, JL, Gregory, CW, Ford, OH, 3rd, et al. The androgen axis in recurrent prostate cancer. Clinical Cancer Research 2004;10:440–8.CrossRef
Montgomery, RB, Mostaghel, EA, Vessella, R, et al. Maintenance of intratumoral androgens in metastatic prostate cancer: a mechanism for castration-resistant tumor growth. Cancer Research 2008;68:4447–54.CrossRef
Dehm, SM, Schmidt, LJ, Heemers, HV, Vessella, RL, Tindall, DJ. Splicing of a novel androgen receptor exon generates a constitutively active androgen receptor that mediates prostate cancer therapy resistance. Cancer Research 2008;68:5469–77.CrossRef
Guo, Z, Yang, X, Sun, F, et al. A novel androgen receptor splice variant is up-regulated during prostate cancer progression and promotes androgen depletion-resistant growth. Cancer Research 2009;69:2305–13.CrossRef
Dehm, SM, Regan, KM, Schmidt, LJ, Tindall, DJ. Selective role of an NH2-terminal WxxLF motif for aberrant androgen receptor activation in androgen depletion independent prostate cancer cells. Cancer Research 2007;67:10 067–77.
Dehm, SM, Tindall, DJ. Ligand-independent androgen receptor activity is activation function-2-independent and resistant to antiandrogens in androgen refractory prostate cancer cells. Journal of Biological Chemistry 2006;281:27 882–93.CrossRefGoogle ScholarPubMed
Narla, G, Difeo, A, Reeves, HL, et al. A germline DNA polymorphism enhances alternative splicing of the KLF6 tumor suppressor gene and is associated with increased prostate cancer risk. Cancer Research 2005;65:1213–22.CrossRef
Narla, G, DiFeo, A, Yao, S, et al. Targeted inhibition of the KLF6 splice variant, KLF6 SV1, suppresses prostate cancer cell growth and spread. Cancer Research 2005;65:5761–8.CrossRef
Chang, B, Zheng, SL, Isaacs, SD, et al. Linkage and association of CYP17 gene in hereditary and sporadic prostate cancer. International Journal of Cancer 2001;95:354–9.3.0.CO;2-3>CrossRefGoogle ScholarPubMed
Alevizaki, M, Cimponeriu, AT, Garofallaki, M, et al. The androgen receptor gene CAG polymorphism is associated with the severity of coronary artery disease in men. Clinical Endocrinology (Oxford) 2003;59:749–55.CrossRef
Canale, D, Caglieresi, C, Moschini, C, et al. Androgen receptor polymorphism (CAG repeats) and androgenicity. Clinical Endocrinology (Oxford) 2005;63:356–61.CrossRef
Lichtenstein, P, Holm, NV, Verkasalo, PK, et al. Environmental and heritable factors in the causation of cancer: analyses of cohorts of twins from Sweden, Denmark, and Finland. New England Journal of Medicine 2000;343:78–85.CrossRefGoogle Scholar
Ahlbom, A, Lichtenstein, P, Malmstrom, H, et al. Cancer in twins: genetic and nongenetic familial risk factors. Journal of the National Cancer Institute 1997;89:287–93.CrossRefGoogle ScholarPubMed
Schaid, DJ, McDonnell, SK, Blute, ML, Thibodeau, SN.Evidence for autosomal dominant inheritance of prostate cancer. American Journal of Human Genetics 1998;62:1425–38.CrossRefGoogle ScholarPubMed
Damber, JE, Aus, G. Prostate cancer. Lancet 2008;371:1710–21.CrossRef
Langeberg, WJ, Isaacs, WB, Stanford, JL. Genetic etiology of hereditary prostate cancer. Frontiers in Bioscience 2007;12:4101–10.CrossRef
Smith, JR, Freije, D, Carpten, JD, et al. Major susceptibility locus for prostate cancer on chromosome 1 suggested by a genome-wide search. Science 1996;274:1371–4.CrossRef
Dong, X, Wang, L, Taniguchi, K, et al. Mutations in CHEK2 associated with prostate cancer risk. American Journal of Human Genetics 2003;72:270–80.CrossRefGoogle ScholarPubMed
Zheng, L, Wang, F, Qian, C, et al. Unique substitution of CHEK2 and TP53 mutations implicated in primary prostate tumors and cancer cell lines. Human Mutation 2006;27:1062–3.CrossRef
Xu, J, Zheng, SL, Komiya, A, et al. Germline mutations and sequence variants of the macrophage scavenger receptor 1 gene are associated with prostate cancer risk. Nature Genetics 2002;32:321–5.CrossRef
Zheng, SL, Sun, J, Wiklund, F, et al. Cumulative association of five genetic variants with prostate cancer. New England Journal of Medicine 2008;358:910–19.CrossRefGoogle ScholarPubMed
Tindall, DJ, Rittmaster, RS. The rationale for inhibiting 5alpha-reductase isoenzymes in the prevention and treatment of prostate cancer. Journal of Urology 2008;179:1235–42.CrossRefGoogle ScholarPubMed
Thompson, IM, Goodman, PJ, Tangen, CM, et al. The influence of finasteride on the development of prostate cancer. New England Journal of Medicine 2003;349:215–24.CrossRefGoogle ScholarPubMed
Thompson, IM, Tangen, C, Goodman, P.The Prostate Cancer Prevention Trial: design, status, and promise. World Journal of Urology 2003;21:28–30.Google ScholarPubMed
Attar, RM, Takimoto, CH, Gottardis, MM. Castration-resistant prostate cancer: locking up the molecular escape routes. Clinical Cancer Research 2009;15:3251–5.CrossRef
Knudsen, KE, Scher, HI. Starving the addiction: new opportunities for durable suppression of AR signaling in prostate cancer. Clinical Cancer Research 2009;15:4792–8.CrossRef
Scher, HI, Sawyers, CL.Biology of progressive, castration-resistant prostate cancer: directed therapies targeting the androgen-receptor signaling axis. Journal of Clinical Oncology 2005;23:8253–61.CrossRefGoogle ScholarPubMed
Tran, C, Ouk, S, Clegg, NJ, et al. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science 2009;324:787–90.CrossRef
Michael, A, Syrigos, K, Pandha, H. Prostate cancer chemotherapy in the era of targeted therapy. Prostate Cancer and Prostatic Diseases 2009;12:13–16.CrossRef

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

  • Prostate cancer
    • By Kaustubh Datta, Department of Urologic Research, Biochemistry and Molecular Biology, Mayo Clinic Cancer Center, Mayo Clinic Foundation, Rochester, MN, USA, Donald J. Tindall, Department of Urologic Research, Biochemistry and Molecular Biology, Mayo Clinic Cancer Center, Mayo Clinic Foundation, Rochester, MN, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.053
Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

  • Prostate cancer
    • By Kaustubh Datta, Department of Urologic Research, Biochemistry and Molecular Biology, Mayo Clinic Cancer Center, Mayo Clinic Foundation, Rochester, MN, USA, Donald J. Tindall, Department of Urologic Research, Biochemistry and Molecular Biology, Mayo Clinic Cancer Center, Mayo Clinic Foundation, Rochester, MN, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.053
Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

  • Prostate cancer
    • By Kaustubh Datta, Department of Urologic Research, Biochemistry and Molecular Biology, Mayo Clinic Cancer Center, Mayo Clinic Foundation, Rochester, MN, USA, Donald J. Tindall, Department of Urologic Research, Biochemistry and Molecular Biology, Mayo Clinic Cancer Center, Mayo Clinic Foundation, Rochester, MN, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.053
Available formats
×