МОЛЕКУЛЯРНО-БИОЛОГИЧЕСКИЕ ФАКТОРЫ ПРОГНОЗА ПРИ РАКЕ ПРЕДСТАТЕЛЬНОЙ ЖЕЛЕЗЫ
https://doi.org/10.21294/1814-4861-2017-16-1-82-90
Аннотация
Цель исследования – обобщить имеющиеся данные о различных молекулярно-генетических диагностических и прогностических маркерах рака предстательной железы. Материал и методы. Поиск соответствующих источников производился в системах Medline, Cochrane Library, Elibrary. Из 540 найденных исследований 61 были использованы для написания систематического обзора. результаты. В настоящее время существует многообразие как прогностических, так и диагностических маркеров, используемых при изучении, диагностике и лечении рака предстательной железы. В обзоре приведена их классификация в зависимости от метода и среды, в которой они были выделены. Рассмотрены молекулярные механизмы участия различных генов и белков в патогенезе и прогрессировании карциномы предстательной железы, представлено потенциальное значение использования их в клинической практике. Заключение. Многие из существующих маркеров могут быть использованы для скрининга и раннего выявления опухоли, а также имеют доказанную прогностическую ценность. Однако противоречивые результаты исследований в отношении некоторых белков и генов требуют дальнейшего изучения, их валидации с последующим внедрением в клиническую практику.
Об авторах
С. В. ВторушинРоссия
доктор медицинских наук, доцент, старший научный сотрудник отделения патологической анатомии и цитологии, Научно-исследовательский институт онкологии, Томский национальный исследовательский медицинский центр Российской академии наук; профессор кафедры патологической анатомии, Сибирский государственный медицинский университет Минздрава России
SPIN-код: 2442-4720.
Н. В. Безгодова
Россия
очный аспирант кафедры патологической анатомии, Сибирский государственный медицинский университет Минздрава России
А. А. Плешкунов
Россия
студент 6-го курса лечебного факультета, Сибирский государственный медицинский университет Минздрава России
Список литературы
1. Чиссов В.И., Старинский В.В., Петрова Г.В. Злокачественные новообразования в России в 2013 году (заболеваемость и смертность). М., 2014; 250.
2. Siegel R., Naishadham D., Jemal A. Cancer Statistics 2013 CA Cancer J Clin. 2013 Jan; 63 (1): 11–30. doi: 10.3322/caac.21166.
3. Cross D.S., Ritter M., Reding D.J. Historical prostate cancer screening and treatment outcomes from a single institution. Clin Med Res. 2012 Aug; 10 (3): 97–105. doi: 10.3121/cmr.2011.1042.
4. Thompson I.M., Pauler D.K., Goodman P.J., Tangen C.M., Lucia M.S., Parnes H.L., Minasian L.M., Ford L.G., Lippman S.M., Crawford E.D., Crowley J.J., Coltman C.A. Jr. Prevalence of prostate cancer among men with a prostate-specific antigen level < or =4.0 ng per milliliter. N Engl J Med. 2004 May 27; 350 (22): 2239–46.
5. Cheng L., Koch M.O., Juliar B.E., Daggy J.K., Foster R.S., Bihrle R., Gardner T.A. The Combined percentage of Gleason patterns 4 and 5 is the best predictor of cancer progression after radical prostatectomy. J Clin Oncol. 2005 May 1; 23 (13): 2911–7.
6. Poncet N., Guillaume J., Mouchiroud G. Epidermal growth factor receptor trans-activation is implicated in IL-6-induced proliferation and ERK1/2 activation in non-transformed prostate epithelial cells. Cell Signal. 2011 Mar; 23 (3): 572–8. doi: 10.1016/j.cellsig.2010.11.009.
7. Guo Y., Xu F., Lu T., Duan Z., Zhang Z. Interleukin-6 signaling pathway in targeted therapy for cancer. Cancer Treat Rev. 2012 Nov; 38 (7): 904–10. doi: 10.1016/j.ctrv.2012.04.007.
8. Nguyen D.P., Li J., Tewari A.K. Inflammation and prostate cancer: the role of interleukin-6. BJU Int. 2014 Jun; 113 (6): 986–92. doi: 10.1111/ bju.12452.
9. Reis S.T., Timoszczuk L.S., Pontes-Junior J., Viana N., Silva I.A., Dip N., Srougi M., Leite K.R. The role of micro RNAs let7c, 100 and 218 expression and their target RAS, C-MYC, BUB1, RB, SMARCA5, LAMB3 and Ki-67 in prostate cancer. Clinics (Sao Paulo). 2013 May; 68 (5): 652–7. doi: 10.6061/clinics/2013(05)12.
10. Lee S.O., Pinder E., Chun J.Y., Lou W., Sun M., Gao A.C. Interleukin-4 stimulates androgen-independent growth in LNCaP human prostate cancer cells. Prostate. 2008 Jan 1; 68 (1): 85–91. doi: 10.1002/pros.20691.
11. Todaro M., Lombardo Y., Francipane M.G., Alea M.P., Cammareri P., Iovino F., Di Stefano A.B., Di Bernardo C., Agrusa A., Condorelli G., Walczak H., Stassi G. Apoptosis resistance in epithelial tumors is mediated by tumor-cell-derived interleukin-4. Cell Death Differ. 2008 Apr; 15 (4): 762–72. doi: 10.1038/sj.cdd.4402305.
12. Takeshi U., Sadar M.D., Suzuki H., Akakura K., Sakamoto S., Shimbo M., Suyama T., Imamoto T., Komiya A., Yukio N., Ichikawa T. Interleukin-4 in patients with prostate cancer Anticancer Res. 2005 NovDec; 25 (6C): 4595–8.
13. Paner G.P., Luthringer D.J., Amin M.B. Best practice in diagnostic immunohistochemistry: prostate carcinoma and its mimics in needle core biopsies. Arch Pathol Lab Med. 2008 Sep; 132 (9): 1388–96. doi: 10.1043/1543-2165(2008)132[1388:BPIDIP]2.0.CO;2.
14. Ma Z., Tsuchiya N., Yuasa T., Huang M., Obara T., Narita S., Horikawa Y., Tsuruta H., Saito M., Satoh S., Ogawa O., Habuchi T. Clinical significance of polymorphism and expression of chromogranin-A and endothelin-1 in prostate cancer. J Urol. 2010 Sep; 184 (3): 1182–8. doi: 10.1016/j.juro.2010.04.063
15. Hong S.K. Kallikreins as Biomarkers for Prostate Cancer. Biomed Res Int. 2014; 2014: 526341. doi: 10.1155/2014/526341.
16. Sardana G., Dowell B., Diamandis E.P. Emerging biomarkers for the diagnosis and prognosis of prostate cancer. Clin Chem. 2008 Dec; 54 (12): 1951–60. doi: 10.1373/clinchem.2008.110668.
17. McCabe N.P., Angwafo F.F. III, Zaher A., Selman S.H., Kouinche A., Jankun J. Expression of soluble urokinase plasminogen activator receptor may be related to outcome in prostate cancer patients. Oncol Rep. 2000 Jul-Aug; 7 (4): 879–82.
18. Shariat S.F., Roehrborn C.G., McConnell J.D., Park S., Alam N., Wheeler T.M., Slawin K.M. Association of the circulating levels of the urokinase system of plasminogen activation with the presence of prostate cancer and invasion, progression, and metastasis. J. Clin. Oncol. 2007; 25: 349–355. doi: 10.1200/JCO.2006.05.6853.
19. Endl E., Gerdes J. The Ki-67 protein: fascinating forms and an unknown function. Exp Cell Res. 2000 Jun 15; 257 (2): 231–7. doi: 10.1006/excr.2000.4888.
20. Tollefson M.K., Karnes R.J., Kwon E.D., Lohse C.M., Rangel L.J., Mynderse L.A., Cheville J.C., Sebo T.J. Prostate Cancer Ki-67 (MIB-1) Expression, Perineural Invasion, and Gleason Score as Biopsy-Based Predictors of Prostate Cancer Mortality: The Mayo Model. Mayo Clin Proc. 2014 Mar; 89 (3): 308–18. doi: 10.1016/j.mayocp.2013.12.001.
21. Miyake H., Muramaki M., Kurahashi T., Takenaka A., Fujisawa M. Expression of potential molecular markers in prostate cancer: correlation with clinicopathological outcomes in patients undergoing radical prostatectomy. Urol Oncol. 2010 Mar-Apr; 28 (2): 145–51. doi: 10.1016/j. urolonc.2008.08.001.
22. Adams J.M., Cory S. Bcl-2-regulated apoptosis: mechanism and therapeutic potential. Curr. Opin. Immunol. 2007; 19: 488–96. doi: 10.1016/j.coi.2007.05.004.
23. Zhou M., Aydin H., Kanane H., Epstein J.I. How often does alphamethylacyl-CoA-racemase contribute to resolving an atypical diagnosis on prostate needle biopsy beyond that provided by basal cell markers? Am. J. Surg. Pathol. 2004; 28: 239–243.
24. Vergis R., Corbishley C.M., Thomas K., Horwich A., Huddart R., Khoo V., Eeles R., Sydes M.R., Cooper C.S., Dearnaley D., Parker C. Expression of Bcl-2, p53, and MDM2 in localized prostate cancer with respect to the outcome of radical radiotherapy dose escalation. Int J Radiat Oncol Biol Phys. 2010 Sep 1; 78 (1): 35–41. doi: 10.1016/j. ijrobp.2009.07.1728.
25. Rennebeck G., Martelli M., Kyprianou N. Anoikis and survival connections in the tumor microenvironment: is there a role is prostate cancer metastasis. Cancer Res. 2005 Dec 15; 65 (24): 11230–5.
26. Kuefer R., Hofer M.D., Zorn C.S., Engel O., Volkmer B.G., JuarezBrito M.A., Eggel M., Gschwend J.E., Rubin M.A., Day M.L. Assessment of a fragment of e-cadherin as a serum biomarker with predictive value for prostate cancer. Br J Cancer. 2005 Jun 6; 92 (11): 2018–23. doi: 10.1038/ sj.bjc.6602599.
27. Uetsuki H., Tsunemori H., Taoka R., Haba R., Ishikawa M., Kakehi Y. Expression of a novel biomarker, EPCA, in adenocarcinomas and precancerous lesions in the prostate. J Urol. 2005 Aug; 174 (2): 514–8. doi: 10.1097/01.ju.0000165154.41159.b1.
28. Dhir R., Vietmeier B., Arlotti J., Acquafondata M., Landsittel D., Masterson R., Getzenberg R.H. Early identification of individuals with prostate cancer in negative biopsies. J Urol. 2004 Apr; 171 (4): 1419–23. doi: 10.1097/01.ju.0000116545.94813.27.
29. Hansel D.E., DeMarzo A.M., Platz E.A., Jadallah S., Hicks J., Epstein J.I., Partin A.W., Netto G.J. Early prostate cancer antigen expression in predicting presence of prostate cancer in men with histologically negative biopsies. J Urol. 2007 May; 177 (5): 1736–40. doi: 10.1016/j. juro.2007.01.013.
30. Leman E.S., Cannon G.W., Trock B.J., Sokoll L.J., Chan D.W., Mangold L., Partin A.W., Getzenberg R.H. EPCA-2: a highly specific serum marker for prostate cancer. Urology. 2007 Apr; 69 (4): 714–20.
31. Ramirez M.L., Nelson E.C., Evans C.P. Beyond prostate-specific antigen: alternate serum markers. Prostate Cancer Prostatic Dis. 2008; 11 (3): 216–29. doi: 10.1038/pcan.2008.2.
32. Han K.R., Seligson D.B., Liu X., Horvath S., Shintaku P.I., Thomas G.V., Said J.W., Reiter R.E. Prostate stem cell antigen expression is associated with Gleason score, seminal vesicle invasion and capsular invasion in prostate cancer. J Urol. 2004 Mar; 171 (3): 1117–21. doi: 10.1097/01. ju.0000109982.60619.93.
33. Gu Z., Thomas G., Yamashiro J., Shintaku I.P., Dorey F., Raitano A., Witte O.N., Said J.W., Loda M., Reiter R.E. Prostate stem cell antigen (PSCA) expression increases with high Gleason score, advanced stage and bone metastasis in prostate cancer. Oncogene. 2000 Mar 2; 19 (10): 1288–96. doi: 10.1038/sj.onc.1203426.
34. Lloyd M.D., Darley D.J., Wierzbicki A.S., Threadgill M.D. Alphamethylacyl-CoA racemase – an ‘obscure’ metabolic enzyme takes centre stage. FEBS J. 2008 Mar; 275 (6): 1089–102. doi: 10.1111/j.1742- 4658.2008.06290.x.
35. Gologan A., Bastacky S., McHale T., Yu J., Cai C., MonzonBordonaba F., Dhir R. Age-associated changes in alpha-methyl CoA racemase (AMACR) expression in nonneoplastic prostatic tissues. Am J Surg Pathol. 2005 Nov; 29 (11): 1435–41.
36. Cardillo M.R., Gentile V., Ceccariello A., Giacomelli L., Messinetti S., Di Silverio F. Can p503s, p504s and p510s gene expression in peripheral-blood be useful as a marker of prostatic cancer? BMC Cancer. 2005; 5: 111. doi: 10.1186/1471-2407-5-111.
37. Ouyang B., Leung Y.K., Wang V., Chung E., Levin L., Bracken B., Cheng L., Ho S.M. Alpha-Methylacyl-CoA racemase spliced variants and their expression in normal and malignant prostate tissues. Urology. 2011 Jan; 77 (1): 249.e1–7. doi: 10.1016/j.urology.2010.08.005.
38. Deshmukh D., Qiu Y. Role of PARP-1 in prostate cancer. Am J Clin Exp Urol. 2015 Apr 25; 3 (1): 1–12.
39. Wacker D.A., Ruhl D.D., Balagamwala E.H., Hope K.M., Zhang T., Kraus W.L. The DNA binding and catalytic domains of poly(ADP-ribose) polymerase-1 cooperate in the regulation of chromatin structure and transcription. Mol Cell Biol. 2007 Nov; 27 (21): 7475–85. doi: 10.1128/ MCB.01314-07.
40. Peralta-Leal A., Rodríguez-Vargas J.M., Aguilar-Quesada R., Rodríguez M.I., Linares J.L., de Almodóvar M.R., Oliver F.J. PARP inhibitors: new partners in the therapy of cancer and inflammatory diseases. Free Radic Biol Med. 2009 Jul 1; 47 (1): 13–26. doi: 10.1016/j. freeradbiomed.2009.04.008.
41. Kumar-Sinha C., Tomlins S.A., Chinnaiyan A.M. Recurrent gene fusions in prostate cancer. Nat Rev Cancer. 2008 Jul; 8 (7): 497–511. doi: 10.1038/nrc2402.
42. Mwamukonda K., Chen Y., Ravindranath L., Furusato B., Hu Y., Sterbis J., Osborn D., Rosner I., Sesterhenn I.A., McLeod D.G., Srivastava S., Petrovics G. Quantitative expression of TMPRSS2 transcript in prostate tumor cells reflects TMPRSS2-ERG fusion status. Prostate Cancer Prostatic Dis. 2010 Mar; 13 (1): 47–51. doi: 10.1038/pcan.2009.28.
43. Tomlins S.A., Rhodes D.R., Perner S., Dhanasekaran S.M., Mehra R., Sun X.W., Varambally S., Cao X., Tchinda J., Kuefer R., Lee C., Montie J.E., Shah R.B., Pienta K.J., Rubin M.A., Chinnaiyan A.M. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science. 2005 Oct 28; 310 (5748): 644–8. doi: 10.1126/science.1117679.
44. Demichelis F., Fall K., Perner S., Andrén O., Schmidt F., Setlur S.R., Hoshida Y., Mosquera J.M., Pawitan Y., Lee C., Adami H.O., Mucci L.A., Kantoff P.W., Andersson S.O., Chinnaiyan A.M., Johansson J.E., Rubin M.A. TMPRSS2:ERG gene fusion associated with lethal prostate cancer in a watchful waiting cohort. Oncogene. 2007 Jul 5; 26 (31): 4596–9. doi: 10.1038/sj.onc.1210237.
45. Chia J.Y., Gajewski J.E., Xiao Y., Zhu H.J., Cheng H.C. Unique biochemical properties of the protein tyrosine phosphatase activity of PTEN-demonstration of different active site structural requirements for phosphopeptide and phospholipid phosphatase activities of PTEN. Biochim Biophys Acta. 2010 Sep; 1804 (9): 1785–95. doi: 10.1016/j. bbapap.2010.05.009.
46. Hessels D., Schalken J.A. Urinary biomarkers for prostate cancer: a review. Asian J Androl. 2013 May; 15 (3): 333–9. doi: 10.1038/ aja.2013.6.
47. Liu L., Yoon J.H., Dammann R., Pfeifer G.P. Frequent hypermethylation of the RASSF1A gene in prostate cancer. Oncogene. 2002 Oct 3; 21 (44): 6835–40. doi: 10.1038/sj.onc.1205814.
48. Kawamoto K., Okino S.T., Place R.F., Urakami S., Hirata H., Kikuno N., Kawakami T., Tanaka Y., Pookot D., Chen Z., Majid S., Enokida H., Nakagawa M., Dahiya R. Epigenetic modifications of RASSF1A gene through chromatin remodeling in prostate cancer. Clin Cancer Res. 2007 May 1; 13 (9): 2541–8. doi: 10.1158/1078-0432.CCR-06-2225.
49. An G., Ng A.Y., Meka C.S., Luo G., Bright S.P., Cazares L., Wright G.L. Jr., Veltri R.W. Cloning and characterization of UROC28, a novel gene overexpressed in prostate, breast, and bladder cancers. Cancer Res. 2000 Dec 15; 60 (24): 7014–20.
50. Pan T., Wu R., Liu B., Wen H., Tu Z., Guo J., Yang J., Shen G. PBOV1 promotes prostate cancer proliferation by promoting G1/S transition. Onco Targets Ther. 2016 Feb 16; 9: 787–95. doi: 10.2147/OTT. S92682.
51. Samusik N., Krukovskaya L., Meln I., Shilov E., Kozlov A. PBOV1 Is a Human De Novo Gene with Tumor-Specific Expression That Is Associated with a Positive Clinical Outcome of Cancer. PLoS One. 2013; 8 (2): e56162. doi: 10.1371/journal.pone.0056162.
52. Lenka G., Weng W.H., Chuang C.K., Ng K.F., Pang S.T. Aberrant expression of the PRAC gene in prostate cancer. Int J Oncol. 2013 Dec; 43 (6): 1960–6. doi: 10.3892/ijo.2013.2117.
53. Norris J.D., Chang C.Y., Wittmann B.M., Kunder R.S., Cui H., Fan D., Joseph J.D., McDonnell D.P. The homeodomain protein HOXB13 regulates the cellular response to androgens. Mol Cell. 2009 Nov 13; 36 (3): 405–16. doi: 10.1016/j.molcel.2009.10.020.
54. Zabalza C.V., Meike A.M., Burdelski C., Wilczak W., Wittmer C., Kraft S., Krech T., Steurer S., Koop C., Hube-Magg C., Graefen M., Heinzer H., Minner S., Simon R., Sauter G., Schlomm T., Tsourlakis M.C. HOXB13 overexpression is an independent predictor of early PSA recurrence in prostate cancer treated by radical prostatectomy. Oncotarget. 2015 May 20; 6 (14): 12822–34. doi: 10.18632/oncotarget.3431.
55. Peng X., Guo W., Liu T., Wang X., Tu X., Xiong D., Chen S., Lai Y., Du H., Chen G., Liu G., Tang Y., Huang S., Zou X. Identification of miRs- 143 and -145 that is associated with bone metastasis of prostate cancer and involved in the regulation of EMT. PLoS One. 2011; 6 (5): e20341. doi: 10.1371/journal.pone.0020341.
56. Li L., Ittmann M.M., Ayala G., Tsai M.J., Amato R.J., Wheeler T.M., Miles B.J., Kadmon D., Thompson T.C. The emerging role of the PI3KAkt pathway in prostate cancer progression. Prostate Cancer Prostatic Dis. 2005; 8(2): 108–18. doi: 10.1038/sj.pcan.4500776.
57. Carnero A., Blanco-Aparicio C., Renner O., Link W., Leal J.F. The PTEN/PI3K/AKT signalling pathway in cancer, therapeutic implications. Curr Cancer Drug Targets. 2008 May; 8 (3): 187–98.
58. Chaux A., Peskoe S.B., Gonzalez-Roibon N., Schultz L., Albadine R., Hicks J., De Marzo A.M., Platz E.A., Netto G.J. Loss of PTEN expression is associated with increased risk of recurrence after prostatectomy for clinically localized prostate cancer. Mod Pathol. 2012 Nov; 25 (11): 1543–9. doi: 10.1038/modpathol.2012.104.
59. Choucair K., Ejdelman J., Brimo F., Aprikian A., Chevalier S., Lapointe J. PTEN genomic deletion predicts prostate cancer recurrence and is associated with low AR expression and transcriptional activity. BMC Cancer. 2012 Nov 22; 12: 543. doi: 10.1186/1471-2407-12-543.
60. Suh S.O., Chen Y., Zaman M.S., Hirata H., Yamamura S., Shahryari V., Liu J., Tabatabai Z.L., Kakar S., Deng G., Tanaka Y., Dahiya R. MicroRNA-145 is regulated by DNA methylation and p53 gene mutation in prostate cancer. Carcinogenesis. 2011 May; 32 (5): 772–8. doi: 10.1093/ carcin/bgr036.
61. Schubert M., Spahn M., Kneitz S., Scholz C.J., Joniau S., Stroebel P., Riedmiller H., Kneitz B. Distinct microRNA expression profile in prostate cancer patients with early clinical failure and the impact of let-7 as prognostic marker in high-risk prostate cancer. PLoS One. 2013 Jun 14; 8 (6): e65064. doi: 10.1371/journal.pone.0065064.
62. Гервас П.А., Литвяков Н.В., Попова Н.О., Добродеев А.Ю., Тарасова А.С., Юмов Е.Л., Иванова Ф.Г., Черемисина О.В., Афанасьев С.Г., Гольдберг В.Е., Чердынцева Н.В. Проблемы и перспективы совершенствования молекулярно-генетической диагностики для назначения таргетных препаратов в онкологии. Сибирский онкологический журнал. 2014; 2: 46–55.
Рецензия
Для цитирования:
Вторушин С.В., Безгодова Н.В., Плешкунов А.А. МОЛЕКУЛЯРНО-БИОЛОГИЧЕСКИЕ ФАКТОРЫ ПРОГНОЗА ПРИ РАКЕ ПРЕДСТАТЕЛЬНОЙ ЖЕЛЕЗЫ. Сибирский онкологический журнал. 2017;16(1):82-90. https://doi.org/10.21294/1814-4861-2017-16-1-82-90
For citation:
Vtorushin S.V., Bezgodova N.V., Pleshkunov A.A. MOLECULAR BIOLOGICAL FACTORS IN THE PREDICTION OF PROSTATE CANCER. Siberian journal of oncology. 2017;16(1):82-90. (In Russ.) https://doi.org/10.21294/1814-4861-2017-16-1-82-90