Hostname: page-component-76fb5796d-22dnz Total loading time: 0 Render date: 2024-04-28T04:22:42.728Z Has data issue: false hasContentIssue false

Review of novel tissue-based biomarkers for prostate cancer: towards personalised and targeted medicine

Published online by Cambridge University Press:  06 April 2021

Ernest Osei*
Affiliation:
Department of Medical Physics, Grand River Regional Cancer Centre, Kitchener, ON, Canada Department of Physics and Astronomy, University of Waterloo, Waterloo, ON, Canada Department of Systems Design Engineering, University of Waterloo, Waterloo, ON, Canada Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
Stephanie Swanson
Affiliation:
Department of Medical Physics, Grand River Regional Cancer Centre, Kitchener, ON, Canada Department of Physics and Astronomy, University of Waterloo, Waterloo, ON, Canada
Lyba Sheraz
Affiliation:
Faculty of Science, School of Interdisciplinary Science, McMaster University, Hamilton, ON, Canada
Nada Khaled Fouad Ibrahim
Affiliation:
Department of Medical Physics, Grand River Regional Cancer Centre, Kitchener, ON, Canada Department of Physics and Astronomy, University of Waterloo, Waterloo, ON, Canada
*
Author for correspondence: Dr Ernest Osei, Department of Medical Physics, Grand River Regional Cancer Centre, 835 King Street West, Kitchener, ON, Canada. Tel: 519 749 4300. E-mail: ernest.osei@grhosp.onca

Abstract

Background:

Prostate cancer is the most commonly diagnosed cancer in men and responsible for about 10% of all cancer mortality in both Canadian and American men. Currently, serum PSA level is the most commonly used test for the detection of prostate cancer, though the levels can also be elevated in benign conditions, has limited specificity and has a high rate of overdiagnosis and treatment of indolent disease. Consequently, in recent years, several investigations have been conducted to identify novel cancer biomarkers capable of both effective screening and diagnosis, as well as assisting to shift the diagnostic and treatment paradigm of prostate cancer towards more patient-specific and targeted medicine. The goal of this narrative review paper is to describe eleven novel and promising tissue-based biomarkers for prostate cancer capable to account for individual patient variabilities and have the potential for risk assessment, early detection and diagnosis, identification of patients who will benefit from a particular treatment and monitoring patient response to treatment.

Materials and methods:

We searched several databases from August to December 2020 for relevant studies published in English between 2000 and 2020 and reporting on tissue-based biomarkers for screening and early diagnosis, treatment and monitoring of prostate cancer.

Conclusions:

Emerging prostate cancer biomarkers have the potential to guide clinical decision-making since they have the potential to detect the disease early, measure the risk of developing the disease and the risk of progression, provide accurate information of patient response to a specific treatment and are capable of informing clinicians about the likely outcome of a cancer diagnosis independent of the treatment received. Therefore, the future holds promise for personalised and targeted medicine from prevention to diagnosis and treatment that considers the individual patient’s variabilities in the management of prostate cancer.

Type
Literature Review
Copyright
© The Author(s), 2021. Published by Cambridge University Press

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

Canadian Cancer Society. Prostate cancer statistics. https://www.cancer.ca/en/cancer-information/cancer-type/prostate/statistics/?region=on. Accessed on 17th April 2020.Google Scholar
Brenner, D R, Weir, H K, Demers, A A et al. Projected estimates of cancer in Canada in 2020. CMAJ 2020; 192 (9): e199e205.CrossRefGoogle ScholarPubMed
Siegel, R L, Miller, K D, Jemal, A. Cancer statistics, 2020. CA Cancer J Clin 2020; 70 (1): 730.CrossRefGoogle ScholarPubMed
White, J, Shenoy, B V, Tutrone, R F et al. Clinical utility of the Prostate Health Index (phi) for biopsy decision management in a large group urology practice setting. Prostate Cancer Prostatic Dis 2017; 21 (1): 7884.CrossRefGoogle Scholar
Osei, E, Swanson, S. A review of current clinical biomarkers for prostate cancer: towards personalised and targeted therapy. J Radiother Pract 2020; 110 (Online Publication).Google Scholar
Kremer, C L, Klein, R R, Mendelson, J et al. Expression of mTOR signaling pathway markers in prostate cancer progression. Prostate 2006; 66 (11): 12031212.CrossRefGoogle ScholarPubMed
Hendriks, R J, van Oort, I M, Schalken, J A. Blood-based and urinary prostate cancer biomarkers: a review and comparison of novel biomarkers for detection and treatment decisions. Prostate Cancer Prostatic Dis 2016; 20 (1): 1219.CrossRefGoogle ScholarPubMed
McGrath, S, Christidis, D, Perera, M et al. Prostate cancer biomarkers: are we hitting the mark? Prostate Int 2016; 4 (4): 130135.CrossRefGoogle Scholar
Guo, J, Yang, J, Zhang, X et al. A panel of biomarkers for diagnosis of prostate cancer using urine samples. Anticancer Res 2018; 38 (3): 14711477.Google ScholarPubMed
Alford, A V, Brito, J M, Yadav, K K, Yadav, S S, Tewari, A K, Renzulli, J. The use of biomarkers in prostate cancer screening and treatment. Rev Urol 2017; 19 (4): 221234.Google ScholarPubMed
Osei, E, Swanson, S, Francis, R. Review of novel liquid-based biomarkers for prostate cancers: towards personalised and targeted medicine. J Radiother Pract 2021 (in Press).Google Scholar
Hessels, D, van Gils, M P, van Hooij, O et al. Predictive value of PCA3 in urinary sediments in determining clinico-pathological characteristics of prostate cancer. Prostate 2010; 70 (1): 1016.CrossRefGoogle ScholarPubMed
Osei, E, Lumini, J, Gunasekara, D et al. A review of predictive, prognostic and diagnostic biomarkers for non-small-cell lung cancer: towards personalised and targeted cancer therapy. J Radiother Pract 2019; 19 (4): 370384.CrossRefGoogle Scholar
Osei, E, Walters, P, Masella, O et al. A review of predictive, prognostic and diagnostic biomarkers for brain tumors: towards personalised and targeted cancer therapy. J Radiother Pract 2019; 116 (Online Publication).Google Scholar
Vanaja, D K, Cheville, J C, Iturria, S J, Young, C Y. Transcriptional silencing of zinc finger protein 185 identified by expression profiling is associated with prostate cancer progression. Cancer Res 2003; 63 (14): 38773882.Google ScholarPubMed
Sánchez-Vega, F, Gotea, V, Petrykowska, H M et al. Recurrent patterns of DNA methylation in the ZNF154, CASP8, and VHL promoters across a wide spectrum of human solid epithelial tumors and cancer cell lines. Epigenetics 2014; 8 (12): 13551372.CrossRefGoogle Scholar
Zhang, W, Shu, P, Wang, S et al. ZNF154 is a promising diagnosis biomarker and predicts biochemical recurrence in prostate cancer. Gene 2018; 675: 136143.CrossRefGoogle ScholarPubMed
Szeliski, K, Adamowicz, J, Gastecka, A, Drewa, T, Pokrywczyńska, M. Modern urology perspectives on prostate cancer biomarkers. Cent European J Urol 2018; 71 (4): 420426.Google ScholarPubMed
Mahapatra, S, Klee, E W, Young, C Y F et al. Global methylation profiling for risk prediction of prostate cancer. Clin Cancer Res 2012; 18 (10): 28822895.CrossRefGoogle ScholarPubMed
Kelly, K A, Setlur, S R, Ross, R et al. Detection of early prostate cancer using a Hepsin-targeted imaging agent. Cancer Res 2008; 68 (7): 22862291.CrossRefGoogle ScholarPubMed
Klezovitch, O, Chevillet, J, Mirosevich, J, Roberts, R L, Matusik, R J, Vasioukhin, V. Hepsin promotes prostate cancer progression and metastasis. Cancer Cell 2004; 6 (2): 185195.CrossRefGoogle ScholarPubMed
Magee, J A, Araki, T, Patil, S et al. Expression profiling reveals Hepsin overexpression in prostate cancer. Cancer Res 2001; 61 (15): 56925696.Google ScholarPubMed
Srikantan, V, Valladares, M, Rhim, J S, Moul, J W, Srivastava, S. HEPSIN inhibits cell growth/invasion in prostate cancer cells. Cancer Res 2002; 62 (23): 68126816.Google ScholarPubMed
Wu, Q, Parry, G. Hepsin and prostate cancer. Front Biosci 2007; 12 (12): 50525059.CrossRefGoogle ScholarPubMed
Tang, X, Mahajan, S S, Nguyen, L T et al. Targeted inhibition of cell-surface serine protease Hepsin blocks prostate cancer bone metastasis. Oncotarget 2014; 5 (5): 13521362.CrossRefGoogle ScholarPubMed
Boss, D S, Beijnen, J H, Schellens, J H M. Clinical experience with aurora kinase inhibitors: a review. Oncologist 2009; 14 (8): 780793.CrossRefGoogle ScholarPubMed
Nigg, E A. Mitotic kinases as regulators of cell division and its checkpoints. Nat Rev Mol Cell Biol 2001; 2 (1): 2132.CrossRefGoogle ScholarPubMed
Lee, E C Y, Frolov, A, Li, R, Ayala, G, Greenberg, N M. Targeting aurora kinases for the treatment of prostate cancer. Cancer Res 2006; 66 (10): 49965002.CrossRefGoogle ScholarPubMed
Kivinummi, K, Urbanucci, A, Leinonen, K et al. The expression of AURKA is androgen regulated in castration-resistant prostate cancer. Sci Rep 2017; 7: 17978.CrossRefGoogle ScholarPubMed
Buschhorn, H M, Klein, R R, Chambers, S M et al. Aurora-A over-expression in high-grade PIN lesions and prostate cancer. Prostate 2005; 64 (4): 341346.CrossRefGoogle ScholarPubMed
Willems, E, Dedobbeleer, M, Digregorio, M, Lombard, A, Lumapat, P N, Rogister, B. The functional diversity of aurora kinases: a comprehensive review. Cell Div 2018; 13: 7.CrossRefGoogle ScholarPubMed
Tang, A, Gao, K, Chu, L, Zhang, R, Yang, J, Zheng, J. Aurora kinases: novel therapy targets in cancers. Oncotarget 2017; 8 (14): 2393723954.CrossRefGoogle ScholarPubMed
Nna, E, Madukwe, J, Egbujo, E et al. Gene expression of aurora kinases in prostate cancer and nodular hyperplasia tissues. Med Princ Pract 2013; 22 (2): 138143.CrossRefGoogle ScholarPubMed
Mankovska, O, Gerashchenko, G, Rozenberg, E et al. Analysis of aurora kinases genes expression points on their distinct roles in prostate cancer development. Ukr Biochem J 2019; 91 (6): 1526.CrossRefGoogle Scholar
Bar-Shira, A, Pinthus, J H, Rozovsky, U et al. Multiple genes in human 20q13 chromosomal region are involved in an advanced prostate cancer xenograft. Cancer Res 2002; 62 (23): 68036807.Google Scholar
Meraldi, P, Honda, R, Nigg, E A. Aurora-A overexpression reveals tetraploidization as a major route to centrosome amplification in p53 −/− cells. EMBO J 2002; 21 (4): 483492.CrossRefGoogle Scholar
Qu, Y, Huang, X, Li, Z et al. miR-199a-3p inhibits aurora kinase A and attenuates prostate cancer growth: new avenue for prostate cancer treatment. Am J Pathol 2014; 184 (5): 15411549.CrossRefGoogle ScholarPubMed
Pihan, G A, Purohit, A, Wallace, J, Malhotra, R, Liotta, L, Doxsey, S J. Centrosome defects can account for cellular and genetic changes that characterize prostate cancer progression. Cancer Res 2001; 61 (5): 22122219.Google ScholarPubMed
Rad, E, Murray, J, Tee, A. Oncogenic signalling through mechanistic target of rapamycin (mTOR): a driver of metabolic transformation and cancer progression. Cancers 2018; 10 (1): 5.CrossRefGoogle ScholarPubMed
Jiang, B, Liu, L. Role of mTOR in anticancer drug resistance: perspectives for improved drug treatment. Drug Resist Updat 2008; 11 (3): 6376.CrossRefGoogle ScholarPubMed
Grunwald, V, DeGraffenried, L, Russel, D, Friedrichs, W E, Ray, R B, Hidalgo, M. Inhibitors of mTOR reverse doxorubicin resistance conferred by PTEN status in prostate cancer cells. Cancer Res 2002; 62 (21): 61416145.Google ScholarPubMed
Forbes, S A, Bindal, N, Bamford, S et al. COSMIC: mining complete cancer genomes in the catalogue of somatic mutations in cancer. Nucleic Acids Res 2010; 39 (suppl 1), D945D950.CrossRefGoogle ScholarPubMed
Sutherland, S I M, Pe Benito, R, Henshall, S M, Horvath, L G, Kench, J G. Expression of phosphorylated-mTOR during the development of prostate cancer. Prostate 2014; 74 (12): 12311239.CrossRefGoogle ScholarPubMed
Brown, R E, Zotalis, G, Zhang, P L, Zhao, B. Morphoproteomic confirmation of a constitutively activated mTOR pathway in high grade prostatic intraepithelial neoplasia and prostate cancer. Int J Clin Exp Pathol 2008; 1 (4): 333342.Google ScholarPubMed
Deeb, D, Gao, X, Jiang, H, Dulchavsky, S A, Gautam, S C. Oleanane Triterpenoid CDDO-Me inhibits growth and induces apoptosis in prostate cancer cells by independently targeting pro-survival Akt and mTOR. Prostate 2009; 69 (8): 851860.CrossRefGoogle ScholarPubMed
Moreno-Bueno, G, Rodríguez-Perales, S, Sánchez-Estévez, C et al. Cyclin D1 gene (CCND1) mutations in endometrial cancer. Oncogene 2003; 22 (38): 61156118.CrossRefGoogle ScholarPubMed
Nakamura, Y, Felizola, S J A, Kurotaki, Y et al. Cyclin D1 (CCND1) expression is involved in estrogen receptor beta (ERβ) in human prostate cancer. Prostate 2013; 73 (6): 590595.CrossRefGoogle Scholar
Casimiro, M C, Di Sante, G, Ju, X et al. Cyclin D1 promotes androgen-dependent DNA damage repair in prostate cancer cells. Cancer Res 2016; 76 (2): 329338.CrossRefGoogle ScholarPubMed
He, Y, Franco, O E, Jiang, M et al. Tissue-specific consequences of cyclin D1 overexpression in prostate cancer progression. Cancer Res 2007; 67 (17): 81888197.CrossRefGoogle ScholarPubMed
Musgrove, E A, Caldon, C E, Barraclough, J, Stone, A, Sutherland, R L. Cyclin D as a therapeutic target in cancer. Nat Rev Cancer 2011; 11 (8): 558572.CrossRefGoogle ScholarPubMed
Peurala, E, Koivunen, P, Haapasaari, K, Bloigu, R, Jukkola-Vuorinen, A. The prognostic significance and value of cyclin D1, CDK4 and p16 in human breast cancer. Breast Cancer Res 2013; 15: R5.CrossRefGoogle ScholarPubMed
Qie, S, Diehl, J A. Cyclin D1, cancer progression, and opportunities in cancer treatment. J Mol Med 2016; 94 (12): 13131326.CrossRefGoogle ScholarPubMed
Tashiro, E, Tsuchiya, A, Imoto, M. Functions of cyclin D1 as an oncogene and regulation of cyclin D1 expression. Cancer Sci 2007; 98 (5): 629635.CrossRefGoogle ScholarPubMed
Drobnjak, M, Osman, I, Scher, H I, Fazzari, M, Cordon-Cardo, C. Overexpression of cyclin D1 is associated with metastatic prostate cancer to bone. Clin Cancer Res 2000; 6 (5): 18911895.Google Scholar
Marampon, F, Gravina, G L, Ju, X et al. Cyclin D1 silencing suppresses tumorigenicity, impairs DNA double strand break repair and thus radiosensitizes androgen-independent prostate cancer cells to DNA damage. Oncotarget 2016; 7 (39): 64526.CrossRefGoogle ScholarPubMed
Comstock, C E S, Revelo, M P, Buncher, C R, Knudsen, K E. Impact of differential cyclin D1 expression and localisation in prostate cancer. Br J Cancer 2007; 96 (6): 970979.CrossRefGoogle ScholarPubMed
Bae, Y K, Kim, A, Kim, M K, Choi, J E, Kang, S H, Lee, S J. Fibronectin expression in carcinoma cells correlates with tumor aggressiveness and poor clinical outcome in patients with invasive breast cancer. Hum Pathol 2013; 44 (10): 20282037.CrossRefGoogle ScholarPubMed
Waalkes, S, Atschekzei, F, Kramer, M W et al. Fibronectin 1 mRNA expression correlates with advanced disease in renal cancer. BMC cancer 2010; 10: 503.CrossRefGoogle ScholarPubMed
Wang, J P, Hielscher, A. Fibronectin: how its aberrant expression in tumors may improve therapeutic targeting. J Cancer 2017; 8 (4): 674682.CrossRefGoogle ScholarPubMed
Williams, C M, Engler, A J, Slone, R D, Galante, L L, Schwarzbauer, J E. Fibronectin expression modulates mammary epithelial cell proliferation during acinar differentiation. Cancer Res 2008; 68 (9): 31853192.CrossRefGoogle ScholarPubMed
Fornaro, M, Plescia, J, Chheang, S et al. Fibronectin protects prostate cancer cells from tumor necrosis factor-α-induced apoptosis via the AKT/Survivin Pathway. J Biol Chem 2003; 278 (50): 5040250411.CrossRefGoogle ScholarPubMed
Sponziello, M, Rosignolo, F, Celano, M et al. Fibronectin-1 expression is increased in aggressive thyroid cancer and favors the migration and invasion of cancer cells. Mol Cell Endocrinol 2016; 431: 123132.CrossRefGoogle ScholarPubMed
Jerhammar, F, Ceder, R, Garvin, S, Grénman, R, Grafström, R, Roberg, K. Fibronectin 1 is a potential biomarker for radioresistance in head and neck squamous cell carcinoma. Cancer Biol Ther 2010; 10 (12): 12441251.CrossRefGoogle ScholarPubMed
Ruiz-Garcia, E, Scott, V, Machavoine, C et al. Gene expression profiling identifies fibronectin 1 and CXCL9 as candidate biomarkers for breast cancer screening. Br J Cancer 2010; 102 (3): 462468.CrossRefGoogle ScholarPubMed
Jia, D, Entersz, I, Butler, C, Foty, R A. Fibronectin matrix-mediated cohesion suppresses invasion of prostate cancer cells. BMC Cancer 2012; 12: 94.CrossRefGoogle ScholarPubMed
Moroz, A, Delella, F K, Lacorte, L M, Deffune, E, Felisbino, S L. Fibronectin induces MMP2 expression in human prostate cancer cells. Biochem Biophys Res Commun 2013; 430 (4): 13191321.CrossRefGoogle ScholarPubMed
Docheva, D, Padula, D, Schieker, M, Clausen-Schaumann, H. Effect of collagen I and fibronectin on the adhesion, elasticity and cytoskeletal organization of prostate cancer cells. Biochem Biophys Res Commun 2010; 402 (2): 361366.CrossRefGoogle ScholarPubMed
Hay, C M, Sult, E, Huang, Q et al. Targeting CD73 in the tumor microenvironment with MEDI9447. Oncoimmunology 2016; 5 (8): e1208875.CrossRefGoogle ScholarPubMed
Ghalamfarsa, G, Kazemi, M H, Mohseni, S R et al. CD73 as a potential opportunity for cancer immunotherapy. Expert Opin Ther Targets 2018; 23 (2): 127142.CrossRefGoogle ScholarPubMed
Zandieh, K, Milani, S, Mohammadi, J, Hashemi, M. Therapeutic value of CD73 as a biomarker in human cancer. J Paramed Sci 2018; 10 (3): 4554.Google Scholar
Yang, Q, Du, J, Zu, L. Overexpression of CD73 in prostate cancer is associated with lymph node metastasis. Pathol Oncol Res 2013; 19 (4): 811814.CrossRefGoogle ScholarPubMed
Wang, L, Tang, S, Wang, Y et al. Ecto-5′-nucleotidase (CD73) promotes tumor angiogenesis. Clin Exp Metastasis 2013; 30 (5): 671680.CrossRefGoogle ScholarPubMed
Beavis, PA, Stagg, J, Darcy, P K, Smyth, M J. CD73: a potent suppressor of antitumor immune responses. Trends Immunol 2012; 33 (5): 231237.CrossRefGoogle ScholarPubMed
Leclerc, B G, Charlebois, R, Chouinard, G et al. CD73 expression is an independent prognostic factor in prostate cancer. Clin Cancer Res 2015; 22 (1): 158166.Google ScholarPubMed
Manser, C, Guillot, F, Vagnoni, A et al. Lemur tyrosine kinase-2 signalling regulates kinesin-1 light chain-2 phosphorylation and binding of Smad2 cargo. Oncogene 2012; 31 (22): 27732782.CrossRefGoogle ScholarPubMed
Eeles, R A, Kote-Jarai, Z, Giles, G G et al. Multiple newly identified loci associated with prostate cancer susceptibility. Nat Genet 2008; 40 (3): 316321.CrossRefGoogle ScholarPubMed
Shah, K, Bradbury, N A. Lemur tyrosine kinase 2, a novel target in prostate cancer therapy. Oncotarget 2015; 6 (16): 1423314246.CrossRefGoogle ScholarPubMed
Conti, A, Majorini, M T, Fontanella, E et al. Lemur tyrosine kinase 2 (LMTK2) is a determinant of cell sensitivity to apoptosis by regulating the levels of the BCL2 family members. Cancer Lett 2016; 389: 5969.CrossRefGoogle ScholarPubMed
Harries, L W, Perry, J R, McCullagh, P, Crundwell, M. Alterations in LMTK2, MSMB and HNF1B gene expression are associated with the development of prostate cancer. BMC Cancer 2010; 10: 315.CrossRefGoogle ScholarPubMed
Kawa, S, Ito, C, Toyama, Y et al. Azoospermia in mice with targeted disruption of the Brek/Lmtk2 (Brain-Enriched Kinase/Lemur Tyrosine Kinase 2) gene. Proc Natl Acad Sci U S A 2006; 103 (51): 1934419349.CrossRefGoogle ScholarPubMed
Chibalina, M V, Seaman, M N J, Miller, C C, Kendrick-Jones, J, Buss, F. Myosin VI and its interacting protein LMTK2 regulate tubule formation and transport to the endocytic recycling compartment. J Cell Sci 2007; 120 (24): 42784288.CrossRefGoogle Scholar
Inoue, T, Kon, T, Ohkura, R et al. BREK/LMTK2 is a myosin VI-binding protein involved in endosomal membrane trafficking. Genes Cells 2008; 13 (5): 483495.CrossRefGoogle ScholarPubMed
Puri, C, Chibalina, M V, Arden, S D, Kruppa, A J, Kendrick-Jones, J, Buss, F. Overexpression of myosin VI in prostate cancer cells enhances PSA and VEGF secretion, but has no effect on endocytosis. Oncogene 2010; 29 (2): 188200.CrossRefGoogle ScholarPubMed
Shui, I M, Lindström, S, Kibel, A S et al. Prostate cancer (PCa) risk variants and risk of fatal PCa in the national cancer institute breast and prostate cancer cohort consortium. Eur Urol 2013; 65 (6): 10691075.CrossRefGoogle Scholar
Heissler, S M, Sellers, J R. Myosins. In: Bradshaw, R A, Stahl, P D (eds). Encyclopedia of Cell Biology. Waltham, USA: Academic Press, 2016: 597607.CrossRefGoogle Scholar
Loikkanen, I, Toljamo, K, Hirvikoski, P, Väisänen, T, Paavonen, T K, Vaarala, M H. Myosin VI is a modulator of androgen-dependent gene expression. Oncol Rep 2009; 22 (5): 991995.Google ScholarPubMed
Dunn, T A, Chen, S, Faith, D A et al. A novel role of myosin VI in human prostate cancer. Am J Pathol 2006; 169 (5): 18431854.CrossRefGoogle ScholarPubMed
Nakamura, K D M, Tilli, T M, Wanderley, J L et al. Osteopontin splice variants expression is involved on docetaxel resistance in PC3 prostate cancer cells. Tumor Biol 2016; 37 (2): 26552663.CrossRefGoogle ScholarPubMed
Aksoy, A, Artas, G, Sevindik, O G. Predictive value of stathmin-1 and osteopontin expression for taxan resistance in metastatic castrate-resistant prostate cancer. Pak J Med Sci 2017; 33 (3): 560565.CrossRefGoogle ScholarPubMed
Forootan, S S, Foster, C S, Aachi, V R et al. Prognostic significance of osteopontin expression in human prostate cancer. Int J Cancer 2006; 118 (9): 22552261.CrossRefGoogle ScholarPubMed
Wisniewski, T, Winiecki, J, Makarewicz, R, Zekanowska, E. The effect of radiotherapy and hormone therapy on osteopontin concentrations in prostate cancer patients. J BUON 2020; 25 (1): 527530.Google Scholar
Weber, G F. The cancer biomarker osteopontin: combination with other markers. Cancer Genomics Proteomics 2011; 8 (6): 263288.Google ScholarPubMed
Weber, G F, Lett, G S, Haubein, N C. Osteopontin is a marker for cancer aggressiveness and patient survival. Brit J Cancer 2010; 103: 861869.CrossRefGoogle ScholarPubMed
Weber, G F, Lett, G S, Haubein, N C. Meta-analysis of osteopontin as a clinical cancer marker. Oncol Rep 2011; 25: 433441.CrossRefGoogle ScholarPubMed
Yuanbing, X U, Gan, X, Wang, Z. Application of osteopontin and Stathmin-1 expression in the evaluation of prostate cancer recurrence and survival prognosis. Chin J Postgrad Med 2018; 41 (7): 642645.Google Scholar
Nagaya, N, Nagata, M, Lu, Y et al. Prostate-specific membrane antigen in circulating tumor cells is a new poor prognostic marker for castration-resistant prostate cancer. PloS One 2020; 15 (1): e0226219.CrossRefGoogle ScholarPubMed
Bouchelouche, K, Choyke, P L, Capala, J. Prostate specific membrane antigen- a target for imaging and therapy with radionuclides. Discov Med 2010; 9 (44): 5561.Google ScholarPubMed
Wüstemann, T, Haberkorn, U, Babich, J, Mier, W. Targeting prostate cancer: prostate-specific membrane antigen based diagnosis and therapy. Med Res Rev 2019; 39 (1): 4069.CrossRefGoogle ScholarPubMed
Paller, C J, Piana, D, Eshleman, J R et al. A pilot study of prostate-specific membrane antigen (PSMA) dynamics in men undergoing treatment for advanced prostate cancer. Prostate 2019; 79 (14): 15971603.CrossRefGoogle ScholarPubMed
Hupe, M C, Philippi, C, Roth, D et al. Expression of prostate-specific membrane antigen (PSMA) on biopsies is an independent risk stratifier of prostate cancer patients at time of initial diagnosis. Front Oncol 2018; 8: 623.CrossRefGoogle ScholarPubMed
Ghosh, A, Heston, W. Understanding prostate-specific membrane antigen and its implication in prostate cancer. In: LaRochelle, W J, Shimkets, R A (eds). The Oncogenomics Handbook. Cancer Drug Discovery and Development. Totowa, NJ: Humana Press, 2005: 597615.Google Scholar
Chang, S S. Overview of prostate-specific membrane antigen. Rev Urol 2004; 6 (suppl 10), S13S18.Google ScholarPubMed
Xiao, Z, Adam, B L, Cazares, L H et al. Quantitation of serum prostate-specific membrane antigen by a novel protein biochip immunoassay discriminates benign from malignant prostate disease. Cancer Res 2001; 61 (16): 60296033.Google ScholarPubMed
Ross, J S, Sheehan, C E, Fisher, H A G et al. Correlation of primary tumor prostate-specific membrane antigen expression with disease recurrence in prostate cancer. Clin Cancer Res 2003; 9 (17): 6357.Google ScholarPubMed
Li, D, Stovall, D B, Wang, W, Sui, G. Advances of zinc signaling studies in prostate cancer. Int J Mol Sci 2020; 21 (2): 667.CrossRefGoogle ScholarPubMed
Tommerup, N, Vissing, H. Isolation and fine mapping of 16 novel human zinc finger-encoding cDNAs identify putative candidate genes for developmental and malignant disorders. Genomics 1995; 27 (2): 259264.CrossRefGoogle ScholarPubMed
Urrutia, R. KRAB-containing zinc-finger repressor proteins. Genome Biol 2003; 4 (10): 231.CrossRefGoogle ScholarPubMed