Междисциплинарные проблемы в онкоурологии


DOI: https://dx.doi.org/10.18565/urology.2019.4(Suppl.).19-24

А.Д. Каприн, Б.Я. Алексеев, А.С. Калпинский

МНИОИ им. П. А. Герцена – филиал ФГБУ НМИРЦ Минздрава России, Москва, Россия
Мультидисциплинарный подход в настоящее время – это необходимый и стандартный подход к лечению онкологических больных. К основным целям мультидисциплинарного подхода относят скоординированное высокоэффективное взаимодействие медицинских специалистов для своевременного выявления, назначения и проведения запланированного объема лечения, а также профилактику и коррекцию побочных эффектов лечения для достижения наиболее продолжительного эффекта от проводимого лечения. В статье обсуждается роль команды мультидисциплинарных специалистов, включившей онколога, уролога, патоморфолога, генетика, радиолога, химиотерапевта, лучевого терапевта, нейрохирурга, ортопеда, рентгенэндоваскулярных, торакальных и абдоминальных хирургов для эффективного лечения онкоурологических больных. Для решения существующих проблем необходимо создать единые стандарты лечения онкоурологических заболеваний, разработать и усовершенствовать систему онкологической помощи, улучшить логистику и повысить квалификацию специалистов или подготовить специалистов по требуемому профилю.

Литература


1. Filella X., Gimenez N. Evaluation of

2. Loeb S. Prostate cancer: Prostate Health Index--improving screening in men with family history. Nat Rev Urol 2013;10:497–498.

3. Gittelman M.C., Hertzman B., Bailen J., et al. PCA3 molecular urine test as a predictor of repeat prostate biopsy outcome in men with previous negative biopsies: a prospective multicenter clinical study. J Urol 2013;190:64–69.

4. Vickers A.J., Gupta A., Savage C.J., et al. A panel of kallikrein marker predicts prostate cancer in a large, population-based cohort followed for 15 years without screening. Cancer Epidemiol Biomarkers Prev 2011;20:255– 261.

5. Parekh D.J., Punnen S., Sjoberg D.D., et al. A multi-institutional prospective trial in the USA confirms that the 4Kscore accurately identifies men with high-grade prostate cancer. Eur Urol 2015;68:464–470.

6. Tomlins S.A., Day J.R., Lonigro R.J., et al. Urine TMPRSS2:ERG plus PCA3 for individualized prostate cancer risk assessment. Eur Urol 2016;70:45–53.

7. Bradley L.A., Palomaki G.E., Gutman S., et al. Comparative effectiveness review: prostate cancer antigen 3 testing for the diagnosis and management of prostate cancer. J Urol 2013;190:389–398.

8. Bishoff J.T., Freedland S.J., Gerber L., et al. Prognostic utility of the cell cycle progression score generated from biopsy in men treated with prostatectomy. J Urol 2014;192:409–414

9. Freedland S.J., Gerber L., Reid J., et al. Prognostic utility of cell cycle progression score in men with prostate cancer after primary external beam radiation therapy. Int J Radiat Oncol Biol Phys 2013;86:848–853.

10. Zhao S.G., Chang S.L., Spratt D.E., et al. Development and validation of a 24-gene predictor of response to postoperative radiotherapy in prostate cancer: a matched, retrospective analysis. Lancet Oncol 2016;17:1612– 1620.

11. Spratt D.E., Zhang J., Santiago-Jimenez M., et al. Development and validation of a novel integrated clinical-genomic risk group classification for localized prostate cancer. J Clin Oncol 2018;36:581–590.

12. Stewart G.D., Van Neste L., Delvenne P., et al. Clinical utility of an epigenetic assay to detect occult prostate cancer in histopathologically negative biopsies: results of the MATLOC study. J Urol 2013;189:1110– 1116.

13. Pritchard C.C., Mateo J., Walsh M.F., et al. Inherited DNA-repair gene mutations in men with metastatic prostate cancer. N Engl J Med 2016;375:443–453.

14. Ewing C.M., Ray A.M., Lange E.M., et al. Germline mutations in HOXB13 and prostate-cancer risk. N Engl J Med 2012;366:141–149.

15. Xu J., Lange E.M., Lu L., et al. HOXB13 is a susceptibility gene for prostate cancer: results from the International Consortium for Prostate Cancer Genetics (ICPCG). Hum Genet 2013;132:5–14.

16. Leongamornlert D., Mahmud N., Tymrakiewicz M., et al. Germline BRCA1 mutations increase prostate cancer risk. Br J Cancer 2012;106:1697–1701.

17. Mersch J., Jackson M.A., Park M., et al. Cancers associated with BRCA1 and BRCA2 mutations other than breast and ovarian. Cancer 2015;121: 269–275.

18. Mitra A., Fisher C., Foster C.S., et al. Prostate cancer in male BRCA1 and BRCA2 mutation carriers has a more aggressive phenotype. Br J Cancer 2008;98:502–507.

19. Thorne H., Willems A.J., Niedermayr E., et al. Decreased prostate cancerspecific survival of men with BRCA2 mutations from multiple breast cancer families. Cancer Prev Res (Phila) 2011;4:1002–1010.

20. Bancroft E.K., Page E.C., Castro E, et al. Targeted prostate cancer screening in BRCA1 and BRCA2 mutation carriers: results from the initial screening round of the IMPACT study. Eur Urol 2014;66:489–499.

21. NCCN Guidelines Version 4.2019 Prostate Cancer. Доступно: https:// nccn.org

22. Bastos D.A., Antonarakis E.S. AR-V7 and treatment selection in advanced prostate cancer: are we there yet? Precis Cancer Med. 2018 Sep; 1: 13.

23. Plymate S.R., Sharp A., de Bono J.S. Nuclear Circulating Tumor Cell Androgen Receptor Variant 7 in Castration-Resistant Prostate Cancer: The Devil Is in the Detail. JAMA Oncol. 2018 Sep 1;4(9):1187–1188.

24. Scher H.I., Graf R.P., Schreiber N.A., Jayaram Anuradha, Winquist Eric, McLaughlin Brigit, Lu David, Fleisher Martin, Orr Sarah, Lowes Lori, Anderson Amanda, Wang Yipeng, Dittamore Ryan, Allan A.L., Attard G., Heller G. Assessment of the Validity of Nuclear-Localized Androgen Receptor Splice Variant 7 in Circulating Tumor Cells as a Predictive Biomarker for Castration-Resistant Prostate Cancer. JAMA Oncol. 2018 Sep; 4(9): 1179–1186.

25. McConkey D.J., Choi W. Molecular Subtypes of Bladder Cancer. Curr Oncol Rep. 2018 Aug 20;20(10):77.

26. Eich M.L., Dyrskjot L., Netto G.J. Toward personalized management in bladder cancer: The promise of novel molecular taxonomy. Virchows Arch. 2017;471:271–280.

27. Moch H., Humphrey P.A., Ulbright T.M., Reuter V.E. WHO Classification of Tumours of the Urinary System and Male Genital Organs. 4th ed. IARC Press; Lyon, France: 2016.

28. Knowles M.A., Hurst C.D. Molecular biology of bladder cancer: New insights into pathogenesis and clinical diversity. Nat. Rev. Cancer. 2015;15:25–41.

29. The Cancer Genome Atlas Research Network Comprehensive molecular characterization of urothelial bladder carcinoma. Nature. 2014;507: 315–322.

30. Kim J., Akbani R., Creighton C.J., Lerner S.P., Weinstein J.N., Getz G., Kwiatkowski D.J. Invasive Bladder Cancer: Genomic Insights and Therapeutic Promise.Clin. Cancer Res. 2015;21:4514–4524.

31. Choi W., Porten S., Kim S., Willis D., Plimack E.R., Hoffman-Censits J., Roth B., Cheng T., Tran M., Lee I.L., et al. Identification of distinct basal and luminal subtypes of muscle-invasive bladder cancer with different sensitivities to frontline chemotherapy. Cancer Cell. 2014;25:152–165.

32. Lindgren D., Frigyesi A., Gudjonsson S., Sjodahl G., Hallden C., Chebil G., Veerla S., Ryden T., Mansson W., Liedberg F., et al. Combined gene expression and genomic profiling define two intrinsic molecular subtypes of urothelial carcinoma and gene signatures for molecular grading and outcome. Cancer Res. 2010;70:3463–3472.

33. Lindgren D., Sjodahl G., Lauss M., Staaf J., Chebil G., Lovgren K., Gudjonsson S., Liedberg F., Patschan O., Mansson W., et al. Integrated genomic and gene expression profiling identifies two major genomic circuits in urothelial carcinoma. PLoS ONE. 2012;7:e38863.

34. Kim P.H., Cha E.K., Sfakianos J.P., Iyer G., Zabor E.C., Scott S.N., Ostrovnaya I., Ramirez R., Sun A., Shah R., et al. Genomic predictors of survival in patients with high-grade urothelial carcinoma of the bladder. Eur. Urol. 2015;67:198–201.

35. Rosenberg J.E., Hoffman-Censits J., Powles T., van der Heijden M.S.,Balar А.V., Necchi A., Dawson N., O’Donnell P.H., Balmanoukian A., Loriot Y., et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: A single-arm, multicentre, phase 2 trial. Lancet. 2016;387:1909–1920.

36. Bellmunt J., de Wit R., Vaughn D.J., Fradet Y., Lee J.L., Fong L., Vogelzang N.J., Climent M.A., Petrylak D.P., Choueiri T.K., et al. Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma. N. Engl. J. Med. 2017;376:1015–1026.

37. NCCN Guidelines Version 4.2019 Bladder Cancer. Доступно: https:// nccn.org

38. Dienstmann R., Rodon J., Prat A., Perez-Garcia J., Adamo B., Felip E., Cortes J., Iafrate A.J., Nuciforo P., Tabernero J. Genomic aberrations in the FGFR pathway: opportunities for targeted therapies in solid tumors. Ann Oncol. 2014 Mar;25(3):552–563.

39. Arlene O. Siefker-Radtke, Andrea Necchi, Se Hoon Park, Jesús GarcÃa Donas, Robert A. Huddart, Earle Frederick Burgess, Mark T. Fleming, Arash Rezazadeh, Begona Mellado, Sergei Varlamov, Monika Joshi, Ignacio Duran, Scott T. Tagawa, Anne OHagan, Anjali Narayan Avadhani, Bob Zhong, Peter De Porre, Yohann Loriot. First results from the primary analysis population of the phase 2 study of erdafitinib (ERDA; JNJ42756493) in patients (pts) with metastatic or unresectable urothelial carcinoma (mUC) and FGFR alterations (FGFRalt). J Clin Oncol 36, 2018 (suppl; abstr 4503).

40. Se Hoon Park, Yohann Loriot, Bob Zhong, Syed A. Hussain, Parthiv Jasvant Mahadevia, Peter De Porre, Arlene O. Siefker-Radtke Erdafitinib in highrisk patients (pts) with advanced urothelial carcinoma (UC). J Clin Oncol 37, 2019 (suppl; abstr 4543).

41. Lamb B.W., Tan W.S., Rehman A., et al. Is prebiopsy MRI good enough to avoid prostate biopsy? A cohort study over a 1-year period. Clin Genitourin Cancer 2015;13:512–517.

42. Pokorny M.R., de Rooij M., Duncan E., et al. Prospective study of diagnostic accuracy comparing prostate cancer detection by transrectal ultrasoundguided biopsy versus magnetic resonance (MR) imaging with subsequent MR-guided biopsy in men without previous prostate biopsies. Eur Urol 2014;66:22–29.

43. Serrao E.M., Barrett T., Wadhwa K., et al. Investigating the ability of multiparametric MRI to exclude significant prostate cancer prior to transperineal biopsy. Can Urol Assoc J 2015;9:E853–858.

44. Weaver J.K., Kim E.H., Vetter J.M., et al. Presence of magnetic resonance imaging suspicious lesion predicts Gleason 7 or greater prostate cancer in biopsy-naive patients. Urology 2016;88:119–124.

45. Ahmed H.U., El-Shater Bosaily A., Brown L.C., et al. Diagnostic accuracy of multi-parametric MRI and TRUS biopsy in prostate cancer (PROMIS): a paired validating confirmatory study. Lancet 2017;389: 815–822.

46. Simmons L.A.M., Kanthabalan A. Arya M., et al. The PICTURE study: diagnostic accuracy of multiparametric MRI in men requiring a repeat prostate biopsy. Br J Cancer 2017;116:1159–1165.

47. Rouviere O., Puech P., Renard-Penna R., et al. Use of prostate systematic and targeted biopsy on the basis of multiparametric MRI in biopsy-naive patients (MRI-FIRST): a prospective, multicentre, paired diagnostic study. Lancet Oncol 2019;20:100–109.

48. Barentsz J.O., Richenberg J., Clements R., et al. ESUR prostate MR guidelines 2012. Eur Radiol 2012;22:746–757.

49. EAU Prostate Cancer Guidelines 2019. Доступно: https://uroweb.org/ guideline/prostate-cancer.

50. Korpics, M.C., et al. Concurrent chemotherapy is associated with improved survival in elderly patients with bladder cancer undergoing radiotherapy. Cancer, 2017. 123: 3524.

51. Roach M., 3rd, et al. Short-term neoadjuvant androgen deprivation therapy and external-beam radiotherapy for locally advanced prostate cancer: longterm results of RTOG 8610. J Clin Oncol, 2008. 26: 585. https://www.ncbi. nlm.nih.gov/pubmed/18172188

52. Dy S.M., et al. Evidence-based standards for cancer pain management. J Clin Oncol, 2008. 26: 3879.

53. Hartsell W.F., et al. Randomized trial of short- versus long-course radiotherapy for palliation of painful bone metastases. J Natl Cancer Inst, 2005. 97: 798.

54. Classen J., et al. Radiotherapy for stages IIA/B testicular seminoma: final report of a prospective multicenter clinical trial. J Clin Oncol, 2003. 21: 1101.

55. Carver B.S., et al. Improved clinical outcome in recent years for men with metastatic nonseminomatous germ cell tumors. J Clin Oncol, 2007. 25: 5603.

56. Alt A.L., et al. Survival after complete surgical resection of multiple metastases from renal cell carcinoma. Cancer, 2011. 117: 2873.

57. Brinkmann O.A., et al. The role of residual tumor resection in patients with metastatic renal cell carcinoma and partial remission following immunotherapy. Eur Urol, 2007: 641.

58. Kwak C., et al. Metastasectomy without systemic therapy in metastatic renal cell carcinoma: comparison with conservative treatment. Urol Int, 2007. 79: 145.

59. Lee S.E., et al. Metastatectomy prior to immunochemotherapy for metastatic renal cell carcinoma. Urol Int, 2006. 76: 256.

60. Petralia, et al. Complete metastasectomy is an independent predictor of cancer-specific survival in patients with clinically metastatic renal cell carcinoma. Eur Urol Suppl 2010, 2010: 162.

61. Russo P., et al. Cytoreductive nephrectomy and nephrectomy/complete metastasectomy for metastatic renal cancer. ScientificWorldJournal,2007. 7: 768.

62. Staehler M., et al. Metastasectomy significantly prolongs survival in patients with metastatic renal cancer. Eur Urol Suppl 2009, 2009: 181. 63. Eggener S.E., et al. Risk score and metastasectomy independently impact prognosis of patients with recurrent renal cell carcinoma. J Urol, 2008. 180: 873.


Об авторах / Для корреспонденции


А в т о р д л я с в я з и: А. С. Калпинский – к.м.н., старший научный сотрудник отдела опухолей репродуктивных и мочевыводящих органов МНИОИ им. П. А. Герцена – филиала ФГБУ «НМИЦ радиологии» Минздрава России, Москва, Россия; e-mail: dr.kalpinskiy@gmail.com


Похожие статьи


Бионика Медиа