全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

A Quest to Identify Prostate Cancer Circulating Biomarkers with a Bench-to-Bedside Potential

DOI: 10.1155/2014/321680

Full-Text   Cite this paper   Add to My Lib

Abstract:

Prostate cancer (PCA) is a major health concern in current times. Ever since prostate specific antigen (PSA) was introduced in clinical practice almost three decades ago, the diagnosis and management of PCA have been revolutionized. With time, concerns arose as to the inherent shortcomings of this biomarker and alternatives were actively sought. Over the past decade new PCA biomarkers have been identified in tissue, blood, urine, and other body fluids that offer improved specificity and supplement our knowledge of disease progression. This review focuses on superiority of circulating biomarkers over tissue biomarkers due to the advantages of being more readily accessible, minimally invasive (blood) or noninvasive (urine), accessible for sampling on regular intervals, and easily utilized for follow-up after surgery or other treatment modalities. Some of the circulating biomarkers like PCA3, IL-6, and TMPRSS2-ERG are now detectable by commercially available kits while others like microRNAs (miR-21, -221, -141) and exosomes hold potential to become available as multiplexed assays. In this paper, we will review some of these potential candidate circulating biomarkers that either individually or in combination, once validated with large-scale trials, may eventually get utilized clinically for improved diagnosis, risk stratification, and treatment. 1. Introduction In 2014, more than 200,000 American men will be diagnosed with prostate cancer (PCA). It is the most frequently diagnosed solid tumor and the second leading cause of cancer-related deaths amongst men in the United States. It is estimated to cause 28% of the total number of cancer cases and 10% of the total cancer deaths amongst adult male cancer patients. One in 6 men carries a lifetime risk of a PCA diagnosis [1]. Prostate-specific antigen (PSA) is the biomarker currently being used for PCA. PSA-based screening test has been proved to be a useful prognostic tool. Usually high preoperative values have been related to advanced disease and a poorer clinical outcome. Clinicians initially used PSA for monitoring PCA patients after treatment to detect disease progression, treatment failure, or potential relapse [2] and then subsequently recommended its use for screening purposes [3–5]. Since the late 1980s, the introduction of PSA screening along with digital rectal exam (DRE) and transrectal ultrasound (TRUS) in general clinical practice has led to an increase in the documented incidence of PCA [6, 7]. This trend has been accompanied by an increase in invasive procedures with radical prostatectomy rates

References

[1]  R. Siegel, D. Naishadham, and A. Jemal, “Cancer statistics,” CA Cancer Journal for Clinicians, vol. 63, no. 1, pp. 11–30, 2013.
[2]  T. A. Stamey, N. Yang, A. R. Hay, et al., “Prostate-specific antigen as a serum marker for adenocarcinoma of the prostate,” The New England Journal of Medicine, vol. 317, no. 15, pp. 909–916, 1987.
[3]  W. H. Cooner, B. R. Mosley, C. L. Rutherford Jr. et al., “Prostate cancer detection in a clinical urological practice by ultrasonography, digital rectal examination and prostate specific antigen,” The Journal of Urology, vol. 143, no. 6, pp. 1146–1154, 1990.
[4]  W. J. Catalona, D. S. Smith, T. L. Ratliff et al., “Measurement of prostate-specific antigen in serum as a screening test for prostate cancer,” The New England Journal of Medicine, vol. 324, no. 17, pp. 1156–1161, 1991.
[5]  C. Parkes, N. J. Wald, P. Murphy et al., “Prospective observational study to assess value of prostate specific antigen as screening test for prostate cancer,” British Medical Journal, vol. 311, no. 7016, pp. 1340–1343, 1995.
[6]  A. L. Potosky, E. J. Feuer, and D. L. Levin, “Impact of screening on incidence and mortality of prostate cancer in the United States,” Epidemiologic Reviews, vol. 23, no. 1, pp. 181–186, 2001.
[7]  A. Shibata, J. Ma, and A. S. Whittemore, “Prostate cancer incidence and mortality in the United States and the United Kingdom,” Journal of the National Cancer Institute, vol. 90, no. 16, pp. 1230–1231, 1998.
[8]  A. M. D. Wolf, R. C. Wender, R. B. Etzioni et al., “American Cancer Society guideline for the early detection of prostate cancer: update 2010,” CA Cancer Journal for Clinicians, vol. 60, no. 2, pp. 70–98, 2010.
[9]  G. Draisma, R. Etzioni, A. Tsodikov et al., “Lead time and overdiagnosis in prostate-specific antigen screening: importance of methods and context,” Journal of the National Cancer Institute, vol. 101, no. 6, pp. 374–383, 2009.
[10]  H. G. Welch and W. C. Black, “Overdiagnosis in cancer,” Journal of the National Cancer Institute, vol. 102, no. 9, pp. 605–613, 2010.
[11]  I. M. Thompson, D. P. Ankerst, C. Chi et al., “Operating characteristics of prostate-specific antigen in men with an initial PSA level of 3.0 ng/mL or lower,” Journal of the American Medical Association, vol. 294, no. 1, pp. 66–70, 2005.
[12]  G. L. Andriole, E. D. Crawford, R. L. Grubb III, et al., “Mortality results from a randomized prostate-cancer screening trial,” The New England Journal of Medicine, vol. 360, pp. 1310–1319, 2009.
[13]  F. H. Schr?der, J. Hugosson, M. J. Roobol, et al., “ERSPC Investigators, Screening and prostate-cancer mortality in a randomized European study,” The New England Journal of Medicine, vol. 360, pp. 1320–1328, 2009.
[14]  S.-D. Qiu, C. Y.-F. Young, D. L. Bilhartz et al., “In situ hybridization of prostate specific antigen mRNA in human prostate,” The Journal of Urology, vol. 144, no. 6, pp. 1550–1556, 1990.
[15]  M. K. Brawer, “Prostate-specific antigen: current status,” CA Cancer Journal for Clinicians, vol. 49, no. 5, pp. 264–281, 1999.
[16]  F. H. Schr?der, H. B. Carter, T. Wolters et al., “Early detection of prostate cancer in 2007—part 1: PSA and PSA kinetics,” European Urology, vol. 53, no. 3, pp. 468–477, 2008.
[17]  I. M. Thompson, D. K. Pauler, P. J. Goodman et al., “Prevalence of prostate cancer among men with a prostate-specific antigen level ≤4.0 ng per milliliter,” The New England Journal of Medicine, vol. 350, no. 22, pp. 2239–2321, 2004.
[18]  A. J. Atkinson, W. A. Colburn, V. G. deGruttola et al., “Biomarkers and surrogate endpoints: preferred definitions and conceptual framework,” Clinical Pharmacology and Therapeutics, vol. 69, no. 3, pp. 89–95, 2001.
[19]  R. S. Svatek, C. Jeldres, P. I. Karakiewicz et al., “Pre-treatment biomarker levels improve the accuracy of post-prostatectomy nomogram for prediction of biochemical recurrence,” Prostate, vol. 69, no. 8, pp. 886–894, 2009.
[20]  S. F. Shariat, J. A. Karam, V. Margulis, and P. I. Karakiewicz, “New blood-based biomarkers for the diagnosis, staging and prognosis of prostate cancer,” BJU International, vol. 101, no. 6, pp. 675–683, 2008.
[21]  S. F. Shariat, J. A. Karam, J. Walz et al., “Improved prediction of disease relapse after radical prostatectomy through a panel of preoperative blood-based biomarkers,” Clinical Cancer Research, vol. 14, no. 12, pp. 3785–3791, 2008.
[22]  S. F. Shariat, J. Walz, C. G. Roehrborn et al., “External validation of a biomarker-based preoperative nomogram predicts biochemical recurrence after radical prostatectomy,” Journal of Clinical Oncology, vol. 26, no. 9, pp. 1526–1531, 2008.
[23]  S. F. Shariat, P. I. Karakiewicz, R. Ashfaq et al., “Multiple biomarkers improve prediction of bladder cancer recurrence and mortality in patients undergoing cystectomy,” Cancer, vol. 112, no. 2, pp. 315–325, 2008.
[24]  C. L. Sawyers, “The cancer biomarker problem,” Nature, vol. 452, no. 7187, pp. 548–552, 2008.
[25]  P. Rolan, “The contribution of clinical pharmacology surrogates and models to drug development—a critical appraisal,” British Journal of Clinical Pharmacology, vol. 44, no. 3, pp. 219–225, 1997.
[26]  Committee on Developing Biomarker-Based Tools for Cancer Screening Diagnosis and Treatment, Cancer Biomarkers: The Promises and Challenges of Improving Detection and Treatment, National Academies Press, Washington, DC, USA, 2007.
[27]  A. Sreekumar, L. M. Poisson, T. M. Rajendiran et al., “Metabolomic profiles delineate potential role for sarcosine in prostate cancer progression,” Nature, vol. 457, no. 7231, pp. 799–800, 2009.
[28]  J. Couzin, “Biomarkers: metabolite in urine may point to high-risk prostate cancer,” Science, vol. 323, no. 5916, p. 865, 2009.
[29]  F. Jentzmik, C. Stephan, K. Miller et al., “Sarcosine in urine after digital rectal examination fails as a marker in prostate cancer detection and identification of aggressive tumours,” European Urology, vol. 58, no. 1, pp. 12–18, 2010.
[30]  G. Lucarelli, M. Fanelli, A. M. V. Larocca et al., “Serum sarcosine increases the accuracy of prostate cancer detection in patients with total serum PSA less than 4.0ng/ml,” Prostate, vol. 72, pp. 1611–1621, 2012.
[31]  A. K. Hewavitharana, “Re: Florian Jentzmik, Carsten Stephan, Kurt Miller, et al. Sarcosine in urine after digital rectal examination fails as a marker in prostate cancer detection and identification of aggressive tumours,” European Urology, vol. 58, no. 4, pp. e39–e40, 2010.
[32]  J. A. Schalken, “Is urinary sarcosine useful to identify patients with significant prostate cancer? The trials and tribulations of biomarker development,” European Urology, vol. 58, no. 1, pp. 19–20, 2010.
[33]  H. J. Issaq and T. D. Veenstra, “Is sarcosine a biomarker for prostate cancer?” Journal of Separation Science, vol. 34, no. 24, pp. 3619–3621, 2011.
[34]  J. Luo, S. Zha, W. R. Gage et al., “α-methylacyl-CoA racemase: a new molecular marker for prostate cancer,” Cancer Research, vol. 62, no. 8, pp. 2220–2226, 2002.
[35]  L.-C. Wu, L.-T. Chen, Y.-J. Tsai et al., “Alpha-methylacyl coenzyme A racemase overexpression in gallbladder carcinoma confers an independent prognostic indicator,” Journal of Clinical Pathology, vol. 65, no. 4, pp. 309–314, 2012.
[36]  Z. Jiang, B. A. Woda, C.-L. Wu, and X. J. Yang, “Discovery and clinical application of a novel prostate cancer marker,” American Journal of Clinical Pathology, vol. 122, no. 2, pp. 275–289, 2004.
[37]  P. J. Zielie, J. A. Mobley, R. G. Ebb, Z. Jiang, R. D. Blute, and S. M. Ho, “A novel diagnostic test for prostate cancer emerges from the determination of α-methylacyl-coenzyme A racemase in prostatic secretions,” The Journal of Urology, vol. 172, no. 3, pp. 1130–1133, 2004.
[38]  B. Laxman, D. S. Morris, J. Yu et al., “A first-generation multiplex biomarker analysis of urine for the early detection of prostate cancer,” Cancer Research, vol. 68, no. 3, pp. 645–649, 2008.
[39]  A. Sreekumar, B. Laxman, D. R. Rhodes et al., “Humoral immune response to α-methylacyl-CoA racemase and prostate cancer,” Journal of the National Cancer Institute, vol. 96, no. 11, pp. 834–843, 2004.
[40]  G. Sardana, B. Dowell, and E. P. Diamandis, “Emerging biomarkers for the diagnosis and prognosis of prostate cancer,” Clinical Chemistry, vol. 54, no. 12, pp. 1951–1960, 2008.
[41]  L. D. Truong, D. Kadmon, B. K. McCune, K. C. Flanders, P. T. Scardino, and T. C. Thompson, “Association of transforming growth factor-β1 with prostate cancer: an immunohistochemical study,” Human Pathology, vol. 24, no. 1, pp. 4–9, 1993.
[42]  V. Ivanovic, A. Melman, B. Davis-Joseph, M. Valcic, and J. Geliebter, “Elevated plasma levels of TGF-β1 in patients with invasive prostate cancer,” Nature Medicine, vol. 1, no. 4, pp. 282–284, 1995.
[43]  S. F. Shariat, M. Shalev, A. Menesses-Diaz et al., “Preoperative plasma levels of transforming growth factor beta1 (TGF-β1 strongly predict progression in patients undergoing radical prostatectomy,” Journal of Clinical Oncology, vol. 19, no. 11, pp. 2856–2864, 2001.
[44]  S. F. Shariat, M. W. Kattan, E. Traxel, et al., “As-sociation of pre- and postoperative plasma levels of transforming growth factor beta(1) and inter-leukin 6 and its soluble receptor with prostate can-cer progression,” Clinical Cancer Research, vol. 10, pp. 1992–1999, 2004.
[45]  S. Akira, T. Taga, and T. Kishimoto, “Interleukin-6 in biology and medicine,” Advances in Immunology, vol. 54, pp. 1–78, 1993.
[46]  A. Hobisch, I. E. Eder, T. Putz et al., “Interleukin-6 regulates prostate-specific protein expression in prostate carcinoma cells by activation of the androgen receptor,” Cancer Research, vol. 58, no. 20, pp. 4640–4645, 1998.
[47]  D. Giri, M. Ozen, and M. Ittmann, “Interleukin-6 is an autocrine growth factor in human prostate cancer,” American Journal of Pathology, vol. 159, no. 6, pp. 2159–2165, 2001.
[48]  V. Michalaki, K. Syrigos, P. Charles, and J. Waxman, “Serum levels of IL-6 and TNF-α correlate with clinicopathological features and patient survival in patients with prostate cancer,” British Journal of Cancer, vol. 90, no. 12, pp. 2312–2316, 2004.
[49]  J. Nakashima, M. Tachibana, Y. Horiguchi et al., “Serum interleukin 6 as a prognostic factor in patients with prostate cancer,” Clinical Cancer Research, vol. 6, no. 7, pp. 2702–2706, 2000.
[50]  M. W. Kattan, S. F. Shariat, B. Andrews et al., “The addition of interleukin-6 soluble receptor and transforming growth factor beta1 improves a preoperative nomogram for predicting biochemical progression in patients with clinically localized prostate cancer,” Journal of Clinical Oncology, vol. 21, no. 19, pp. 3573–3579, 2003.
[51]  R. H. Getzenberg, K. J. Pienta, E. Y. W. Huang, and D. S. Coffey, “Identification of nuclear matrix proteins in the cancer and normal rat prostate,” Cancer Research, vol. 51, no. 24, pp. 6514–6520, 1991.
[52]  H. Uetsuki, H. Tsunemori, R. Taoka, R. Haba, M. Ishikawa, and Y. Kakehi, “Expression of a novel biomarker, EPCA, in adenocarcinomas and precancerous lesions in the prostate,” The Journal of Urology, vol. 174, no. 2, pp. 514–518, 2005.
[53]  R. Dhir, B. Vietmeier, J. Arlotti et al., “Early identification of individuals with prostate cancer in negative biopsies,” The Journal of Urology, vol. 171, no. 4, pp. 1419–1423, 2004.
[54]  B. Paul, R. Dhir, D. Landsittel, M. R. Hitchens, and R. H. Getzenberg, “Detection of prostate cancer with a blood-based assay for early prostate cancer antigen,” Cancer Research, vol. 65, no. 10, pp. 4097–4100, 2005.
[55]  Z. Zhao, W. Ma, G. Zeng, D. Qi, L. Ou, and Y. Liang, “Preoperative serum levels of early prostate cancer antigen (EPCA) predict prostate cancer progression in patients undergoing radical prostatectomy,” Prostate, vol. 72, no. 3, pp. 270–279, 2012.
[56]  M. J. G. Bussemakers, A. van Bokhoven, G. W. Verhaegh et al., “DD3: a new prostate-specific gene, highly overexpressed in prostate cancer,” Cancer Research, vol. 59, no. 23, pp. 5975–5979, 1999.
[57]  J. B. de Kok, G. W. Verhaegh, R. W. Roelofs et al., “DD3PCA3, a very sensitive and specific marker to detect prostate tumors,” Cancer Research, vol. 62, no. 9, pp. 2695–2698, 2002.
[58]  H. Nakanishi, J. Groskopf, H. A. Fritsche et al., “PCA3 molecular urine assay correlates with prostate cancer tumor volume: implication in selecting candidates for active surveillance,” The Journal of Urology, vol. 179, no. 5, pp. 1804–1810, 2008.
[59]  E. J. Whitman, J. Groskopf, A. Ali et al., “PCA3 score before radical prostatectomy predicts extracapsular extension and tumor volume,” The Journal of Urology, vol. 180, no. 5, pp. 1975–1978, 2008.
[60]  A. Haese, A. de la Taille, H. van Poppel et al., “Clinical utility of the PCA3 urine assay in European men scheduled for repeat biopsy,” European Urology, vol. 54, no. 5, pp. 1081–1088, 2008.
[61]  L. S. Marks, Y. Fradet, I. Lim Deras et al., “PCA3 molecular urine assay for prostate cancer in men undergoing repeat biopsy,” Urology, vol. 69, no. 3, pp. 532–535, 2007.
[62]  F. K. Chun, A. de la Taille, H. van Poppel, et al., “Prostate cancer gene 3 (PCA3): development and internal validation of a novel biopsy nomogram,” European Urology, vol. 56, pp. 659–668, 2009.
[63]  B. Ouyang, B. Bracken, B. Burke, E. Chung, J. Liang, and S.-M. Ho, “A duplex quantitative polymerase chain reaction assay based on quantification of alpha-methylacyl-CoA racemase transcripts and prostate cancer antigen 3 in urine sediments improved diagnostic accuracy for prostate cancer,” The Journal of Urology, vol. 181, no. 6, pp. 2508–2513, 2009.
[64]  G. Kristiansen, F. R. Fritzsche, K. Wassermann et al., “GOLPH2 protein expression as a novel tissue biomarker for prostate cancer: implications for tissue-based diagnostics,” British Journal of Cancer, vol. 99, no. 6, pp. 939–948, 2008.
[65]  Y. Mao, H. Yang, H. Xu et al., “Golgi protein 73 (GOLPH2) is a valuable serum marker for hepatocellular carcinoma,” Gut, vol. 59, no. 12, pp. 1687–1693, 2010.
[66]  F. Zhang, Y. Gu, X. Li, W. Wang, J. He, and T. Peng, “Up-regulated Golgi phosphoprotein 2 (GOLPH2) expression in lung adenocarcinoma tissue,” Clinical Biochemistry, vol. 43, no. 12, pp. 983–991, 2010.
[67]  J. R. Prensner and A. M. Chinnaiyan, “Oncogenic gene fusions in epithelial carcinomas,” Current Opinion in Genetics and Development, vol. 19, no. 1, pp. 82–91, 2009.
[68]  S. A. Tomlins, D. R. Rhodes, S. Perner et al., “Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer,” Science, vol. 310, no. 5748, pp. 644–648, 2005.
[69]  S. A. Tomlins, S. M. J. Aubin, J. Siddiqui et al., “Urine TMPRSS2: ERG fusion transcript stratifies prostate cancer risk in men with elevated serum PSA,” Science Translational Medicine, vol. 3, no. 94, Article ID 94ra72, 2011.
[70]  D. Hessels, F. P. Smit, G. W. Verhaegh, J. A. Witjes, E. B. Cornel, and J. A. Schalken, “Detection of TMPRSS2-ERG fusion transcripts and prostate cancer antigen 3 in urinary sediments may improve diagnosis of prostate cancer,” Clinical Cancer Research, vol. 13, no. 17, pp. 5103–5108, 2007.
[71]  G. Bahrenberg, A. Brauers, H.-G. Joost, and G. Jakse, “Reduced expression of PSCA, a member of the LY-6 family of cell surface antigens, in bladder, esophagus, and stomach tumors,” Biochemical and Biophysical Research Communications, vol. 275, no. 3, pp. 783–788, 2000.
[72]  Z. Gu, G. Thomas, J. Yamashiro et al., “Prostate stem cell antigen (PSCA) expression increases with high gleason score, advanced stage and bone metastasis in prostate cancer,” Oncogene, vol. 19, no. 10, pp. 1288–1296, 2000.
[73]  A. G. de Nooij-van Dalen, G. A. M. S. van Dongen, S. J. Smeets et al., “Characterization of the human Ly-6 antigens, the newly annotated member Ly-6K included, as molecular markers for head-and-neck squamous cell carcinoma,” International Journal of Cancer, vol. 103, no. 6, pp. 768–774, 2003.
[74]  M. L. Beckett, L. H. Cazares, A. Vlahou, P. F. Schellhammer, and G. L. Wright Jr., “Prostate-specific membrane antigen levels in sera from healthy men and patients with benign prostate hyperplasia or prostate cancer,” Clinical Cancer Research, vol. 5, no. 12, pp. 4034–4040, 1999.
[75]  R. E. Reiter, Z. Gu, T. Watabe et al., “Prostate stem cell antigen: a cell surface marker overexpressed in prostate cancer,” Proceedings of the National Academy of Sciences of the United States of America, vol. 95, no. 4, pp. 1735–1740, 1998.
[76]  K.-R. Han, D. B. Seligson, X. Liu et al., “Prostate stem cell antigen expression is associated with gleason score, seminal vesicle invasion and capsular invasion in prostate cancer,” The Journal of Urology, vol. 171, no. 3, pp. 1117–1121, 2004.
[77]  M. L. Ramírez, E. C. Nelson, and C. P. Evans, “Beyond prostate-specific antigen: alternate serum markers,” Prostate Cancer and Prostatic Diseases, vol. 11, no. 3, pp. 216–229, 2008.
[78]  S. Isshiki, K. Akakura, A. Komiya, H. Suzuki, N. Kamiya, and H. Ito, “Chromogranin a concentration as a serum marker to predict prognosis after endocrine therapy for prostate cancer,” The Journal of Urology, vol. 167, no. 2, pp. 512–515, 2002.
[79]  N. Hara, T. Kasahara, T. Kawasaki et al., “Reverse transcription-polymerase chain reaction detection of prostate-specific antigen, prostate-specific membrane antigen, and prostate stem cell antigen in one milliliter of peripheral blood: value for the staging of prostate cancer,” Clinical Cancer Research, vol. 8, no. 6, pp. 1794–1799, 2002.
[80]  S. Ross, S. D. Spencer, I. Holcomb et al., “Prostate stem cell antigen as therapy target: tissue expression and in vivo efficacy of an immunoconjugate,” Cancer Research, vol. 62, no. 9, pp. 2546–2553, 2002.
[81]  Z. Gu, J. Yamashiro, E. Kono, and R. E. Reiter, “Anti-prostate stem cell antigen monoclonal antibody 1G8 induces cell death in vitro and inhibits tumor growth in vivo via a Fc-independent mechanism,” Cancer Research, vol. 65, no. 20, pp. 9495–9500, 2005.
[82]  D. C. Saffran, A. B. Raitano, R. S. Hubert, O. N. Witte, R. E. Reiter, and A. Jakobovits, “Anti-PSCA mAbs inhibit tumor growth and metastasis formation and prolong the survival of mice bearing human prostate cancer xenografts,” Proceedings of the National Academy of Sciences of the United States of America, vol. 98, no. 5, pp. 2658–2663, 2001.
[83]  J. Dannull, P.-A. Diener, L. Prikler et al., “Prostate stem cell antigen is a promising candidate for immunotherapy of advanced prostate cancer,” Cancer Research, vol. 60, no. 19, pp. 5522–5528, 2000.
[84]  J. V. Tricoli, M. Schoenfeldt, and B. A. Conley, “Detection of prostate cancer and predicting progression: current and future diagnostic markers,” Clinical Cancer Research, vol. 10, no. 12, pp. 3943–3953, 2004.
[85]  D. P. Bartel, “MicroRNAs: genomics, biogenesis, mechanism, and function,” Cell, vol. 116, no. 2, pp. 281–297, 2004.
[86]  G. A. Calin and C. M. Croce, “MicroRNA signatures in human cancers,” Nature Reviews Cancer, vol. 6, no. 11, pp. 857–866, 2006.
[87]  C. M. Croce, “Causes and consequences of microRNA dysregulation in cancer,” Nature Reviews Genetics, vol. 10, no. 10, pp. 704–714, 2009.
[88]  Y. Pang, C. Y. F. Young, and H. Yuan, “MicroRNAs and prostate cancer,” Acta Biochimica et Biophysica Sinica, vol. 42, no. 6, pp. 363–369, 2010.
[89]  F. Y. Agaoglu, M. Kovancilar, Y. Dizdar et al., “Investigation of miR-21, miR-141, and miR-221 in blood circulation of patients with prostate cancer,” Tumor Biology, vol. 32, no. 3, pp. 583–588, 2011.
[90]  R. Medina, S. K. Zaidi, C.-G. Liu et al., “MicroRNAs 221 and 222 bypass quiescence and compromise cell survival,” Cancer Research, vol. 68, no. 8, pp. 2773–2780, 2008.
[91]  N. Mercatelli, V. Coppola, D. Bonci et al., “The inhibition of the highly expressed mir-221 and mir-222 impairs the growth of prostate carcinoma xenografts in mice,” PLoS ONE, vol. 3, no. 12, Article ID e4029, 2008.
[92]  L. Wang, H. Tang, V. Thayanithy et al., “Gene networks and microRNAs implicated in aggressive prostate cancer,” Cancer Research, vol. 69, no. 24, pp. 9490–9497, 2009.
[93]  G.-H. Shi, D.-W. Ye, X.-D. Yao et al., “Involvement of microRNA-21 in mediating chemo-resistance to docetaxel in androgen-independent prostate cancer PC3 cells,” Acta Pharmacologica Sinica, vol. 31, no. 7, pp. 867–873, 2010.
[94]  Z. Lu, M. Liu, V. Stribinskis et al., “MicroRNA-21 promotes cell transformation by targeting the programmed cell death 4 gene,” Oncogene, vol. 27, no. 31, pp. 4373–4379, 2008.
[95]  J. Shen, G. W. Hruby, J. M. Mckiernan et al., “Dysregulation of circulating microRNAs and prediction of aggressive prostate cancer,” Prostate, vol. 72, pp. 1469–1477, 2012.
[96]  A. W. Tong, P. Fulgham, C. Jay et al., “MicroRNA profile analysis of human prostate cancers,” Cancer Gene Therapy, vol. 16, no. 3, pp. 206–216, 2009.
[97]  M. Spahn, S. Kneitz, C.-J. Scholz et al., “Expression of microRNA-221 is progressively reduced in aggressive prostate cancer and metastasis and predicts clinical recurrence,” International Journal of Cancer, vol. 127, no. 2, pp. 394–403, 2010.
[98]  E. S. Martens-Uzunova, S. E. Jalava, N. F. Dits et al., “Diagnostic and prognostic signatures from the small non-coding RNA transcriptome in prostate cancer,” Oncogene, vol. 31, no. 8, pp. 978–991, 2012.
[99]  R. J. Bryant, T. Pawlowski, J. W. F. Catto et al., “Changes in circulating microRNA levels associated with prostate cancer,” British Journal of Cancer, vol. 106, no. 4, pp. 768–774, 2012.
[100]  H. C. Nguyen, W. Xie, M. Yang, et al., “Expression differences of circulating microRNAs in metastatic castration resistant prostate cancer and low-risk, localized prostate cancer,” Prostate, vol. 73, pp. 346–354, 2013.
[101]  D. Kong, Y. Li, Z. Wang et al., “miR-200 regulates PDGF-D-mediated epithelial-mesenchymal transition, adhesion, and invasion of prostate cancer cells,” Stem Cells, vol. 27, no. 8, pp. 1712–1721, 2009.
[102]  X. Peng, W. Guo, T. Liu et al., “Identification of miRs-143 and -145 that is associated with bone metastasis of prostate cancer and involved in the regulation of EMT,” PLoS ONE, vol. 6, no. 5, Article ID e20341, 2011.
[103]  S. Rani, K. O'Brien, F. C. Kelleher et al., “Isolation of exosomes for subsequent mRNA, MicroRNA, and protein profiling,” Methods in Molecular Biology, vol. 784, pp. 181–195, 2011.
[104]  G. Tavoosidana, G. Ronquist, S. Darmanis et al., “Multiple recognition assay reveals prostasomes as promising plasma biomarkers for prostate cancer,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 21, pp. 8809–8814, 2011.
[105]  D. Duijvesz, T. Luider, C. H. Bangma, and G. Jenster, “Exosomes as biomarker treasure chests for prostate cancer,” European Urology, vol. 59, no. 5, pp. 823–831, 2011.
[106]  J. Nilsson, J. Skog, A. Nordstrand et al., “Prostate cancer-derived urine exosomes: a novel approach to biomarkers for prostate cancer,” British Journal of Cancer, vol. 100, no. 10, pp. 1603–1607, 2009.
[107]  P. J. Mitchell, J. Welton, J. Staffurth et al., “Can urinary exosomes act as treatment response markers in prostate cancer?” Journal of Translational Medicine, vol. 7, article 4, 2009.
[108]  J. Sandoval and M. Esteller, “Cancer epigenomics: beyond genomics,” Current Opinion in Genetics and Development, vol. 22, no. 1, pp. 50–55, 2012.
[109]  L. A. Deroo, S. C. Bolick, Z. Xu, et al., “Global DNA methylation and one-carbon metabolism gene polymorphisms and the risk of breast cancer in the Sister Study,” Carcinogenesis, 2013.
[110]  Y. Mori, K. Cai, Y. Cheng et al., “A genome-wide search identifies epigenetic silencing of somatostatin, tachykinin-1, and 5 other genes in colon cancer,” Gastroenterology, vol. 131, no. 3, pp. 797–808, 2006.
[111]  K. Kron, V. Pethe, L. Briollais et al., “Discovery of novel hypermethylated genesin prostate cancer using genomic CpG island microarrays,” PLoS ONE, vol. 4, no. 3, Article ID e4830, 2009.
[112]  T. A. Chan and S. B. Baylin, “Epigenetic biomarkers,” Current Topics in Microbiology and Immunology, vol. 355, pp. 189–216, 2011.
[113]  R. Holliday, “The inheritance of epigenetic defects,” Science, vol. 238, no. 4824, pp. 163–170, 1987.
[114]  A. P. Feinberg and B. Vogelstein, “Hypomethylation distinguishes genes of some human cancers from their normal counterparts,” Nature, vol. 301, no. 5895, pp. 89–92, 1983.
[115]  W. Goering, M. Kloth, and W. A. Schulz, “DNA methylation changes in prostate cancer,” Methods in Molecular Biology, vol. 863, pp. 47–66, 2012.
[116]  A. S. Perry, R. W. G. Watson, M. Lawler, and D. Hollywood, “The epigenome as a therapeutic target in prostate cancer,” Nature Reviews Urology, vol. 7, no. 12, pp. 668–680, 2010.
[117]  D. Takai and P. A. Jones, “Comprehensive analysis of CpG islands in human chromosomes 21 and 22,” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 6, pp. 3740–3745, 2002.
[118]  Y. Wang and F. C. C. Leung, “An evaluation of new criteria for CpG islands in the human genome as gene markers,” Bioinformatics, vol. 20, no. 7, pp. 1170–1177, 2004.
[119]  A. Bird, “The essentials of DNA methylation,” Cell, vol. 70, no. 1, pp. 5–8, 1992.
[120]  M. Esteller, “Molecular origins of cancer: epigenetics in cancer,” The New England Journal of Medicine, vol. 358, no. 11, pp. 1148–1096, 2008.
[121]  M. Nakayama, C. J. Bennett, J. L. Hicks et al., “Hypermethylation of the human glutathione S-transferase-π gene (GSTP1) CpG island is present in a subset of proliferative inflammatory atrophy lesions but not in normal or hyperplastic epithelium of the prostate: a detailed study using laser-capture microdissection,” American Journal of Pathology, vol. 163, no. 3, pp. 923–933, 2003.
[122]  M. O. Hoque, O. Topaloglu, S. Begum et al., “Quantitative methylation-specific polymerase chain reaction gene patterns in urine sediment distinguish prostate cancer patients from control subjects,” Journal of Clinical Oncology, vol. 23, no. 27, pp. 6569–6575, 2005.
[123]  M. L. Gonzalgo, C. P. Pavlovich, S. M. Lee, and W. G. Nelson, “Prostate cancer detection by GSTP1 methylation analysis of postbiopsy urine specimens,” Clinical Cancer Research, vol. 9, no. 7, pp. 2673–2677, 2003.
[124]  C. Jerónimo, H. Usadel, R. Henrique et al., “Quantitative GSTP1 hypermethylation in bodily fluids of patients with prostate cancer,” Urology, vol. 60, no. 6, pp. 1131–1135, 2002.
[125]  M. Rouprêt, V. Hupertan, D. R. Yates et al., “Molecular detection of localized prostate cancer using quantitative methylation-specific PCR on urinary cells obtained following prostate massage,” Clinical Cancer Research, vol. 13, no. 6, pp. 1720–1725, 2007.
[126]  J. Ellinger, K. Haan, L. C. Heukamp et al., “CpG island hypermethylation in cell-free serum DNA identifies patients with localized prostate cancer,” Prostate, vol. 68, no. 1, pp. 42–49, 2008.
[127]  C. Chao, M. Chi, M. Preciado, et al., “Methylation markers for prostate cancer prognosis: a systematic review,” Cancer Causes Control, vol. 24, pp. 1615–1641, 2013.
[128]  T. R. Ashworth, “A case of cancer in which cells similar to those in the tumours were seen in the blood after death,” Australasian Medical Journal, vol. 14, pp. 146–147, 1869.
[129]  T. M. Gorges and K. Pantel, “Circulating tumor cells as therapy-related biomarkers in cancer patients,” Cancer Immunology, Immunotherapy, vol. 62, no. 5, pp. 931–939, 2013.
[130]  M.-Y. Kim, T. Oskarsson, S. Acharyya et al., “Tumor self-seeding by circulating cancer cells,” Cell, vol. 139, no. 7, pp. 1315–1326, 2009.
[131]  G. Attard, J. F. Swennenhuis, D. Olmos et al., “Characterization of ERG, AR and PTEN gene status in circulating tumor cells from patients with castration-resistant prostate cancer,” Cancer Research, vol. 69, no. 7, pp. 2912–2918, 2009.
[132]  D. C. Danila, G. Heller, G. A. Gignac et al., “Circulating tumor cell number and prognosis in progressive castration-resistant prostate cancer,” Clinical Cancer Research, vol. 13, no. 23, pp. 7053–7058, 2007.
[133]  J. S. de Bono, H. I. Scher, R. B. Montgomery et al., “Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer,” Clinical Cancer Research, vol. 14, no. 19, pp. 6302–6309, 2008.
[134]  R. Suades, T. Padro, G. Vilahur, et al., “Circulating and platelet-derived microparticles in human blood enhance thrombosis on atherosclerotic plaques,” Thrombosis and Haemostasis, vol. 108, pp. 1208–1219, 2012.
[135]  B. A. Kerr, R. Miocinovic, A. K. Smith, E. A. Klein, and T. V. Byzova, “Comparison of tumor and microenvironment secretomes in plasma and in platelets during prostate cancer growth in a xenograft model,” Neoplasia, vol. 12, no. 5, pp. 388–396, 2010.
[136]  R. J. A. Nilsson, L. Balaj, E. Hulleman et al., “Blood platelets contain tumor-derived RNA biomarkers,” Blood, vol. 118, no. 13, pp. 3680–3683, 2011.
[137]  K.-I. Lin, N. Chattopadhyay, M. Bai et al., “Elevated extracellular calcium can prevent apoptosis via the calcium-sensing receptor,” Biochemical and Biophysical Research Communications, vol. 249, no. 2, pp. 325–331, 1998.
[138]  G. G. Schwartz, “Is serum calcium a biomarker of fatal prostate cancer?” Future Oncology, vol. 5, no. 5, pp. 577–580, 2009.
[139]  H. G. Skinner and G. G. Schwartz, “Serum calcium and incident and fatal prostate cancer in the national health and nutrition examination survey,” Cancer Epidemiology Biomarkers and Prevention, vol. 17, no. 9, pp. 2302–2305, 2008.
[140]  J. L. Peterson, S. J. Buskirk, M. G. Heckman, et al., “Evaluation of serum calcium as a predictor of biochemical recurrence following salvage radiation therapy for prostate cancer,” ISRN Oncology, vol. 2013, Article ID 239241, 7 pages, 2013.
[141]  J. W. Dennis and M. Granovsky, “Protein glycosylation in development and disease,” Bioassays, vol. 21, no. 5, pp. 412–421, 1999.
[142]  J. B. Lowe and J. D. Marth, “A genetic approach to mammalian glycan function,” Annual Review of Biochemistry, vol. 72, pp. 643–691, 2003.
[143]  M. L. A. de Leoz, H. J. An, S. Kronewitter et al., “Glycomic approach for potential biomarkers on prostate cancer: profiling of N-linked glycans in human sera and pRNS cell lines,” Disease Markers, vol. 25, no. 4-5, pp. 243–258, 2008.
[144]  S.-I. Hakomori, “Tumor malignancy defined by aberrant glycosylation and sphingo(glyco)lipid metabolism,” Cancer Research, vol. 56, no. 23, pp. 5309–5318, 1996.
[145]  A. Kobata, “A retrospective and prospective view of glycopathology,” Glycoconjugate Journal, vol. 15, no. 4, pp. 323–331, 1998.
[146]  Z. Kyselova, Y. Mechref, M. M. Al Bataineh et al., “Alterations in the serum glycome due to metastatic prostate cancer,” Journal of Proteome Research, vol. 6, no. 5, pp. 1822–1832, 2007.
[147]  R. Saldova, Y. Fan, J. M. Fitzpatrick, R. W. G. Watson, and P. M. Rudd, “Core fucosylation and α2-3 sialylation in serum N-glycome is significantly increased in prostate cancer comparing to benign prostate hyperplasia,” Glycobiology, vol. 21, no. 2, pp. 195–205, 2011.
[148]  A. Sarrats, R. Saldova, J. Comet et al., “Glycan characterization of PSA 2-DE subforms from serum and seminal plasma,” OMICS A Journal of Integrative Biology, vol. 14, no. 4, pp. 465–474, 2010.
[149]  A. Sarrats, J. Comet, G. Tabarés et al., “Differential percentage of serum prostate-specific antigen subforms suggests a new way to improve prostate cancer diagnosis,” Prostate, vol. 70, no. 1, pp. 1–9, 2010.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133