-
1
-
2Academic Journal
Authors: Vladimir A. Vorobev, Andrey V. Shcherbatykh, Kirill B. Lelyavin, Artur R. Tukhiev, Kirill M. Su-Yanz, Asiya I. Syrova, Amina I. Syrova
Source: Байкальский медицинский журнал, Vol 4, Iss 2, Pp 22-31 (2025)
Subject Terms: рак предстательной железы, метастазы в глаз, офтальмологические метастазы, хориоидальные метастазы, орбитальные метастазы, пэт/кт с psma, андроген-депривационная терапия, Medicine (General), R5-920
File Description: electronic resource
-
3Academic Journal
-
4Academic Journal
Authors: V. A. Andreev, A. V. Nezvetskiy, Yu. D. Udalov, V. A. Kiselev, S. E. Gritsenko, I. V. Nezvetskaya, В. А. Андреев, А. В. Незвецкий, Ю. В. Удалов, В. А. Киселев, С. Е. Гриценко, И. В. Незвецкая
Source: Diagnostic radiology and radiotherapy; Том 16, № 3 (2025); 115-120 ; Лучевая диагностика и терапия; Том 16, № 3 (2025); 115-120 ; 2079-5343
Subject Terms: протонная терапия, prostate cancer, rectal cancer, proton therapy, рак предстательной железы, рак прямой кишки
File Description: application/pdf
Relation: https://radiag.bmoc-spb.ru/jour/article/view/1151/710; Зубеев П.С., Коровин О.А., Коновалов В.А. и др. Случай эффективной диагностики и лечения рака толстой кишки с прорастанием в предстательную железу // Современные технологии в медицине. 2012. № 2. С. 148–150.; Козлова И.В., Удалов Ю.Д., Данилова Л.А. и др. Протонная лучевая терапия рака прямой кишки: литературный обзор // Тазовая хирургия и онкология. 2023. Т. 13, № 1. С. 33–38. doi:10.17650/2686-9594-2023-13-1-33-38.; Незвецкая И.В., Удалов Ю.Д., Незвецкий А.В. и др. Результаты применения протонной лучевой терапии в лечении рака прямой кишки // Колопроктология. 2024. Т. 23, № 2 (88). С. 61–67. doi:10.33878/2073-7556-2024-23-2-61-67.; Ткачев С.И., Булычкин П.В., Матвеев В.Б. и др. Спасительная лучевая терапия рецидивов рака предстательной железы после радикальной простатэктомии // Онкоурология. 2018. Т. 14, № 1. С. 100–106. doi:10.17650/1726-9776-2018-14-1-100-106.; Малихов А.Г., Барсуков Ю.А., Ткачев С.И. и др. Рак прямой кишки: некоторые аспекты // Вестник РОНЦ им. Н. Н. Блохина РАМН. 2015. Т. 26, № 2. С. 25– 29.; Bryant C.M., Henderson R.H., Nichols R.C. et al. Genitourinary Subcommittee of the Particle Therapy Co-Operative Group. Consensus Statement on Proton Therapy for Prostate Cancer // Int. J. Part. Ther. 2021. Apr. Vol. 12, No. 8 (2). P. 1–16. doi:10.14338/IJPT-20-00031.; Trofimov A., Nguyen P.L., Coen J.J. et al. Radiotherapy treatment of early-stage prostate cancer with IMRT and protons: a treatment planning comparison // Int. J. Radiat. Oncol. Biol. Phys. 2007. Oct. Vol. 1, No. 69 (2). P. 444–453. doi:10.1016/j.ijrobp.2007.03.018.; Chera B.S., Vargas C., Morris C.G. et al. Dosimetric study of pelvic proton radiotherapy for high-risk prostate cancer // Int. J. Radiat. Oncol. Biol. Phys. 2009. Nov. Vol. 15, No. 75 (4). P. 994–1002. doi:10.1016/j.ijrobp.2009.01.044.
-
5Academic Journal
Authors: T. Yu. Kochetova, V. V. Krylov, S. A. Ivanov, A. D. Kaprin, Т. Ю. Кочетова, В. В. Крылов, С. А. Иванов, А. Д. Каприн
Source: Siberian journal of oncology; Том 23, № 6 (2024); 32-40 ; Сибирский онкологический журнал; Том 23, № 6 (2024); 32-40 ; 2312-3168 ; 1814-4861
Subject Terms: общая выживаемость, 225 Ac-PSMA-617, castrate-resistant prostate cancer, radium chloride [ 223 Ra], overall survival, 225 Ас-ПСМА-617, кастрационно-резистентный рак предстательной железы, радия хлорид [223Ra]
File Description: application/pdf
Relation: https://www.siboncoj.ru/jour/article/view/3360/1304; Sartor O., de Bono J., Chi K.N., Fizazi K., Herrmann K., Rahbar K., Tagawa S.T., Nordquist L.T., Vaishampayan N., El-Haddad G., Park C.H., Beer T.M., Armour A., Pérez-Contreras W.J., DeSilvio M., Kpamegan E., Gericke G., Messmann R.A., Morris M.J., Krause B.J.; VISION Investigators. Lutetium-177-PSMA-617 for Metastatic Castration-Resistant Prostate Cancer. N Engl J Med. 2021; 385(12): 1091–1103. doi:10.1056/NEJMoa2107322.; Kratochwil C., Bruchertseifer F., Rathke H., Bronzel M., Apostolidis C., Weichert W., Haberkorn U., Giesel F.L., Morgenstern A. Targeted α-Therapy of Metastatic Castration-Resistant Prostate Cancer with 225Ac-PSMA-617: Dosimetry Estimate and Empiric Dose Finding. J Nucl Med. 2017; 58(10): 1624–31. doi:10.2967/jnumed.117.191395.; Sathekge M.M., Lawal I.O., Bal C., Bruchertseifer F., Ballal S., Cardaci G., Davis C., Eiber M., Hekimsoy T., Knoesen O., Kratochwil C., Lenzo N.P., Mahapane J., Maserumule L.C., Mdlophane A.H., Mokoala K.M.G., Ndlovu H., Pant V., Rathke H., Reed J., Sen I.B., Singh A., Sood A., Tauber R., Thakral P., Yadav M.P., Morgenstern A. Actinium-225-PSMA radioligand therapy of metastatic castration-resistant prostate cancer (WARMTH Act): a multicentre, retrospective study. Lancet Oncol. 2024; 25(2): 175–83. doi:10.1016/S1470-2045(23)00638-1.; Dai Y.H., Chen P.H., Lee D.J., Andrade G., Vallis K.A. A Meta-Analysis and Meta-Regression of the Efficacy, Toxicity, and Quality of Life Outcomes Following Prostate-Specific Membrane Antigen Radioligand Therapy Utilising Lutetium-177 and Actinium-225 in Metastatic Prostate Cancer. Eur Urol. 2024. doi:10.1016/j.eururo.2024.09.020.; Майстренко Д.Н., Станжевский А.А., Важенина Д.А., Одинцова М.В., Попов С.А., Номоконова В.Б., Чипига Л.А., Сапрыкин К.А., Громов А.В., Васильев С.К. Радиолигандная терапия препаратами на основе радионуклида 225Ас: опыт Российского научного центра радиологии и хирургических технологий имени академика А.М. Гранова. Лучевая диагностика и терапия. 2022; 13(4): 86–94. doi:10.22328/2079-5343-2022-13-4-86-94.; Кочетова Т.Ю., Крылов В.В., Сигов М.А., Рипп В.О., Шуринов А.Ю., Васильев К.Г., Легкодимова Н.С., Иванов С.А., Каприн А.Д. Пилотное исследование безопасности трех возрастающих активностей 225Ас-ПСМА для лечения метастатического кастрационно-резистентного рака предстательной железы. Онкологический журнал: лучевая диагностика, лучевая терапия. 2024; 7(1): 30–40. doi:10.37174/2587-7593-2024-7-1-30-40.; Shiner A., Sperandio R.C., Naimi M., Emmenegger U. Prostate Cancer Liver Metastasis: An Ominous Metastatic Site in Need of Distinct Management Strategies. J Clin Med. 2024; 13(3): 734. doi:10.3390/jcm13030734.; Rahbar K., Essler M., Eiber M., la Fougère C., Prasad V., Fendler W.P., Rassek P., Hasa E., Dittmann H., Bundschuh R.A., Pabst K.M., Kurtinecz M., Schmall A., Verholen F., Sartor O. 177Lu-Prostate-Specific Membrane Antigen Therapy in Patients with Metastatic Castration-Resistant Prostate Cancer and Prior 223Ra (RALU Study). J Nucl Med. 2023; 64(12): 1925–31. doi:10.2967/jnumed.123.266125.; https://www.siboncoj.ru/jour/article/view/3360
-
6Academic Journal
Authors: V. M. Perepukhov, B. Ya. Alekseev, K. M. Nyushko, В. М. Перепухов, Б. Я. Алексеев, К. М. Нюшко
Source: Cancer Urology; Том 20, № 4 (2024); 132-138 ; Онкоурология; Том 20, № 4 (2024); 132-138 ; 1996-1812 ; 1726-9776
Subject Terms: исследования реальной клинической практики, prostate cancer, Radium-223 dichloride, Radium-223, radiopharmaceutical, androgen deprivation therapy, median overall survival, median progression-free survival, real-world clinical practice studies, рак предстательной железы, дихлорид радия-223, Радий-223, радиофармацевтический препарат, андрогенная депривационная терапия, медиана общей выживаемости, медиана выживаемости без прогрессирования
File Description: application/pdf
Relation: https://oncourology.abvpress.ru/oncur/article/view/1903/1576; Шахзадова А.О., Старинский В.В., Лисичникова И.В. Состояние онкологической помощи населению России в 2022 году. Сибирский онкологический журнал 2023;22(5):5–13.; Freedland S.J., Humphreys E.B., Mangold L.A. et al. Risk of prostate cancer-specific mortality following biochemical recurrence after radical prostatectomy. JAMA 2005;294(4):433–9. DOI:10.1001/jama.294.4.433; Kupelian P.A., Buchsbaum J.C., Elshaikh M. et al. Factors affecting recurrence rates after prostatectomy or radiotherapy in localized prostate carcinoma patients with biopsy Gleason score 8 or above. Cancer 2002;95(11):2302–7. DOI:10.1002/cncr.10977; Рак предстательной железы. Клинические рекомендации. Минздрав России, 2021.; Gallaher J., Cook L.M., Gupta S. et al. Improving treatment strategies for patients with metastatic castrate resistant prostate cancer through personalized computational modeling. Clin Exp Metastasis 2014;31:991–9. DOI:10.1007/s10585-014-9674-1; Kirby M., Hirst C., Crawford E.D. Characterising the castration-resistant prostate cancer population: a systematic review. Int J Clin Pract 2011;65(11):1180–92. DOI:10.1111/j.1742-1241.2011.02799.x; Bubendorf L., Schöpfer A., Wagner U. et al. Metastatic patterns of prostate cancer: an autopsy study of 1,589 patients. Hum Pathol 2000;31(5):578–83. DOI:10.1053/hp.2000.6698; Smith M., Parker C., Saad F. et al. Addition of radium-223 to abiraterone acetate and prednisone or prednisolone in patients with castration-resistant prostate cancer and bone metastases (ERA 223): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2019;20(3):408–19. DOI:10.1016/S1470-2045(18)30860-X; Parker C., Nilsson S., Heinrich D. et al. Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med 2013;369(3):213–23. DOI:10.1056/NEJMoa1213755; Алексеев Б.Я., Томбаль Б., Кельн А.А. Рак предстательной железы в условиях ограниченных возможностей. Онкоурология 2021;17(4):112–4. DOI:10.17650/1726-9776-2021-17-4-112-114; Gillessen S. Presented at: European Society for Medical Oncology Congress; September 13–17, 2024; Barcelona, Spain. Abstract LBA1.; Maughan B.L., Kessel A., McFarland T.R. et al. Radium-223 plus enzalutamide versus enzalutamide in metastatic castration-refractory prostate cancer: final safety and efficacy results. Oncologist 2021;26(12):1006–e2129. DOI:10.1002/onco.13949; Morris M.J., De Wit R., Vogelzang N.J. et al. A phase III trial of docetaxel versus docetaxel and radium-223 (Ra-223) in patients with metastatic castration-resistant prostate cancer (mCRPC): DORA. J Clin Oncol 2021;39:TPS5091.; Bruland Ø.S., Nilsson S., Fisher D.R., Larsen R.H. et al. High-linear energy transfer irradiation targeted to skeletal metastases by the α-emitter 223Ra: adjuvant or alternative to conventional modalities? Clin Cancer Res 2006;12(20):6250s–7s. DOI:10.1158/1078-0432.CCR-06-0841; Kerr C. (223)Ra targets skeletal metastases and spares normal tissue. Lancet Oncol 2002;3(8):453. DOI:10.1016/s1470-2045(02)00835-5; Hyväkkä A., Kääriäinen O.S., Utriainen T. et al. Radium-223 dichloride treatment in metastatic castration-resistant prostate cancer in Finland: A real-world evidence multicenter study. Cancer Med 2023;12(4):4064–76. DOI:10.1002/cam4.5262; Matsumoto T. Effectiveness and safety of radium-223 dichloride in patients with castration-resistant prostate cancer and bone metastases in real-world practice: a multi-institutional study. Int J Urol 2023;30(2):139–46.; Zhang H. Survival and fracture risk with radium-223 therapy in metastatic castrate-resistant prostate cancer (mCRPC): a real-world analysis. J Clin Oncol 2024;42:50.; Юрмазов З.А., Усынин Е.А., Медведева А.А. и др. Клинический опыт применения радия-223 у больных с костными метастазами при кастрационно-резистентном раке предстательной железы. Онкоурология 2022;18(1):70–6. DOI:10.17650/1726-9776-2022-18-1-70-76.; Van der Doelen M.J., Oving I.M., Wyndaele D.N.J. et al. Health-related quality of life, psychological distress, and fatigue in metastatic castration-resistant prostate cancer patients treated with radium-223 therapy. Prostate Cancer Prostatic Dis 2023;26(1):142–50. DOI:10.1038/s41391-022-00569-8; Кит О.И., Максимова Н.А., Гусарева М.А. и др. Применение внутривенной радионуклидной терапии радия хлоридом [223Ra] у пациентов с костными метастазами кастрационно-резистентного рака предстательной железы. Южно-Российский онкологический журнал 2024;5(4):6–13. DOI:10.37748/2686-9039-2024-5-4-1; Матвеев В.Б., Маркова А.С. Эффективность и безопасность радия-223 в исследованиях реальной клинической практики. Онкоурология 2020;16(4):129–35. DOI:10.17650/1726-9776-2020-16-4-129-135; https://www.pharmjournal.ru/jour/announcement/view/1527; https://prostorpharma.ru/preparaty/rflp-radiya-xlorid-223ra/; https://oncourology.abvpress.ru/oncur/article/view/1903
-
7Academic Journal
Authors: A. V. Sarkisyan, P. V. Bulychkin, A. V. Khachaturyan, A. A. Kufelkina, D. D. Ladyko, А. В. Саркисян, П. В. Булычкин, А. В. Хачатурян, А. А. Куфелкина, Д. Д. Ладыко
Source: Cancer Urology; Том 20, № 4 (2024); 55-59 ; Онкоурология; Том 20, № 4 (2024); 55-59 ; 1996-1812 ; 1726-9776
Subject Terms: стереотаксическая лучевая терапия, metachronous oligometastases, hormone-sensitive prostate cancer, stereotactic radiation therapy, метахронные олигометастазы, гормоночувствительный рак предстательной железы
File Description: application/pdf
Relation: https://oncourology.abvpress.ru/oncur/article/view/1833/1582; Состояние онкологической помощи населению России в 2021 году. Под ред. А.Д. Каприна, В.В. Старинского, А.О. Шахзадовой. М.: МНИОИ им. П.А. Герцена – филиал ФГБУ «НМИЦ радиологии» Минздрава России, 2022. 239 с.; Cornford P., Bellmunt J., Bolla M. et al. EAU-ESTRO-SIOG guidelines on prostate cancer. Part II: treatment of relapsing, metastatic, and castration-resistant prostate cancer. Eur Urol 2017;71(4):630–42. DOI:10.1016/j.eururo.2016.08.002; Hellman S., Weichselbaum R.R. Oligometastases. J Clin Oncol 1995;13(1):8–10. DOI:10.1200/JCO.1995.13.1.8; Phillips R., Yue Shi W., Deek M. et al. Outcomes of observation vs stereotactic ablative radiation for oligometastatic prostate cancer: the ORIOLE phase 2 randomized clinical trial. JAMA Oncol 2020;6(5):650–9. DOI:10.1001/jamaoncol.2020.0147; Palma D.A., Olson R., Harrow S. et al. Stereotactic ablative radiotherapy for the comprehensive treatment of oligometastatic cancers: long-term results of the SABR-COMET phase II randomized trial. J Clin Oncol 2020;38:2830–8. DOI:10.1200/JCO.20.00818; Tang C., Sherry A.D., Haymaker C. et al. Addition of metastasis-directed therapy to intermittent hormone therapy for oligometastatic prostate cancer: the EXTEND phase 2 randomized clinical trial. JAMA Oncol 2023;9(6):825–34. DOI:10.1001/jamaoncol.2023.0161; https://oncourology.abvpress.ru/oncur/article/view/1833
-
8Academic Journal
Authors: A. A. Li, T. M. Geliashvili, A. A. Rumyantsev, A. I. Pronin, A. S. Krylov, O. D. Baranova, A. V. Parnas, V. S. Ilyakov, D. A. Vorobyeva, А. А. Ли, Т. М. Гелиашвили, А. А. Румянцев, А. И. Пронин, А. С. Крылов, О. Д. Баранова, А. В. Парнас, В. С. Ильяков, Д. А. Воробьева
Source: Cancer Urology; Том 20, № 4 (2024); 98-103 ; Онкоурология; Том 20, № 4 (2024); 98-103 ; 1996-1812 ; 1726-9776
Subject Terms: 177 Lu-ПСМА-617, metastatic castration-resistant prostate cancer, 177 Lu-PSMA-617, метастатический кастрационно-резистентный рак предстательной железы
File Description: application/pdf
Relation: https://oncourology.abvpress.ru/oncur/article/view/1818/1587; https://oncourology.abvpress.ru/oncur/article/downloadSuppFile/1818/1514; https://oncourology.abvpress.ru/oncur/article/downloadSuppFile/1818/1515; Bray F., Laversanne M., Sung H. et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2024;74(3):229–63. DOI:10.3322/caac.21834; Шахзадова А.О., Старинский В.В., Лисичникова И.В. Состояние онкологической помощи населению России в 2022 году. Сибирский онкологический журнал 2023;22(5):5–13. DOI:10.21294/1814-4861-2023-22-5-5-13; Состояние онкологической помощи населению России в 2022 году. Под ред. А.Д. Каприна, В.В. Старинского, А.О. Шахзадовой. М.: МНИОИ им. П.А. Герцена – филиал ФГБУ «НМИЦ радиологии» Минздрава России, 2023. 239 с.; Kirby M., Hirst C., Crawford E.D. Characterising the castration-resistant prostate cancer population: a systematic review. Int J Clin Pract 2011;65(11):1180–92. DOI:10.1111/j.1742-1241.2011.02799.x; Hofman M.S., Hicks R.J., Maurer T., Eiber M. Prostate-specific Membrane Antigen PET: Clinical Utility in Prostate Cancer, Normal Patterns, Pearls, and Pitfalls. Radiographics 2018;38(1):200–17. DOI:10.1148/rg.2018170108; Sartor O., de Bono J., Chi K.N. et al.; VISION Investigators. Lutetium-177-PSMA-617 for metastatic castration-resistant prostate cancer. N Engl J Med 2021;385(12):1091–103. DOI:10.1056/NEJMoa2107322; Fallah J., Agrawal S., Gittleman H. et al. FDA Approval Summary: lutetium Lu 177 vipivotide tetraxetan for patients with metastatic castration-resistant prostate cancer. Clin Cancer Res 2023;29(9):1651–7. DOI:10.1158/1078-0432.CCR-22-2875; https://oncourology.abvpress.ru/oncur/article/view/1818
-
9Academic Journal
Authors: M. I. Volkova, Al-I. S. Akel, Ya. V. Gridneva, M. Yu. Fedyanin, I. A. Pokataev, S. G. Vardanyan, R. I. Ryabinin, V. N. Galkin, М. И. Волкова, Аль-И. С. Акел, Я. В. Гриднева, М. Ю. Федянин, И. А. Покатаев, С. Г. Варданян, Р. И. Рябинин, В. Н. Галкин
Source: Malignant tumours; Том 15, № 1 (2025); 36–45 ; Злокачественные опухоли; Том 15, № 1 (2025); 36–45 ; 2587-6813 ; 2224-5057
Subject Terms: отсроченное лечение, non-metastatic prostate cancer, radical prostatectomy, radiation therapy, delayed treatment, неметастатический рак предстательной железы, радикальная простатэктомия, лучевая терапия
File Description: application/pdf
Relation: https://www.malignanttumors.org/jour/article/view/1447/1034; Состояние онкологической помощи населению России в 2022 году. Под ред. А.Д. Каприна, В.В. Старинского, А.О. Шахзадовой 2022. М.: МНИОИ им. П.А. Герцена − филиал ФГБУ «НМИЦ радиологии» Минздрава России, 2022.; Ferlay J., Colombet M., Soerjomataram I., et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer 2019;144(8):1941–53. https://doi.org/10.1002/ijc.31937; Носов Д.А., Волкова М.И., Гладков О.А. и соавт. Практические рекомендации по лекарственному лечению рака предстательной железы. Злокачественные опухоли 2023;13(3s2):640–660. https://doi.org/10.18027/2224-5057-2023-13-3s2-1-640-660.; NCCN guideline for prostate cancer. Retrieved from https://www.nccn.org/home; Van Poppel H., Roobol M.J., Chapple C.R., et al. Prostate-specific antigen testing as part of a risk-adapted early detection strategy for prostate cancer: European Association of Urology Position and Recommendations for 2021. Eur Urol 2021;80(6):703–711. https://doi.org/10.1016/j.eururo.2021.07.024; Hamdy F.C., Donovan J.L., Lane J.A., et al. Fifteen-year outcomes after monitoring, surgery, or radiotherapy for prostate cancer. N Engl J Med 2023;388(17):1547–1558. https://doi.org/10.1056/NEJMoa2214122; Bill-Axelson A., Holmberg L., Garmo H., et al. Radical prostatectomy or watchful waiting in prostate cancer — 29- year follow-up. N Engl J Med 2018;379:2319–2329. https://doi.org/10.1056/NEJMoa1807801; Wilt T.J., Brawer M.K., Jones K.M., et al. Radical prostatectomy versus observation for localized prostate cancer. N Engl J Med 2012;367(3):203–13. https://doi.org/10.1056/NEJMoa1113162; Boyle H.J., Alibhai S., Decoster L., et al. Updated recommendations of the International Society of Geriatric Oncology on prostate cancer management in older patients. Eur J Cancer 2019;116:116–36. https://doi.org/10.1016/j.ejca.2019.04.031; Akre O., Garmo H., Adolfsson J., et al. Mortality among men with locally advanced prostate cancer managed with noncurative intent: a nationwide study in PCBaSe Sweden. Eur Urol 2011;60(3):554–563. https://doi.org/10.1016/j.eururo.2011.05.047; Albertsen P.C., Hanley J.A., Fine J. 20-year outcomes following conservative management of clinically localized prostate cancer. JAMA 2005;293(17):2095–2101. https://doi.org/10.1001/jama.293.17.2095; Bylow K., Mohile S.G., Stadler W. M, Dale W. Does androgen-deprivation therapy accelerate the development of frailty in older men with prostate cancer?: a conceptual review. Cancer 2007;110(12):2604–2613. https://doi.org/10.1002/cncr.23084; Widmark A, Tomic R, Modig H, et al. Prospective randomized trial comparing external beam radiotherapy versus watchful waiting in early prostate cancer (T1b-T2, pN0, grade 1–2, M0). Presented at the 53rd Annual ASTRO Meeting, Miami Beach, FL, October 2–6, 2011; Iversen P., Madsen P.O., Corle D.K. Radical prostatectomy versus expectant treatment for early carcinoma of the prostate. Twenty-three year follow-up of a prospective randomized study. Scand J Urol Nephrol Suppl 1995;172:65–72.; Bill-Axelson A., Holmberg L., Ruutu M., et al. Radical prostatectomy versus watchful waiting in localized prostate cancer. N Engl J Med 2011;364(18):1708–1717. https://doi.org/10.1056/NEJMoa1011967; Van Hemelrijck M., Garmo H., Lindhagen L., et al. Quantifying the transition from active surveillance to watchful waiting among men with very low-risk prostate cancer. Eur Urol 2017;72(4):534–541. https://doi.org/10.1016/j.eururo.2016.10.031.; https://www.malignanttumors.org/jour/article/view/1447
-
10Academic Journal
Authors: M. I. Shaladonova, S. A. Usanov, М. И. Шаладонова, С. А. Усанов
Source: Doklady of the National Academy of Sciences of Belarus; Том 69, № 2 (2025); 117-128 ; Доклады Национальной академии наук Беларуси; Том 69, № 2 (2025); 117-128 ; 2524-2431 ; 1561-8323 ; 10.29235/1561-8323-2025-69-2
Subject Terms: гибкий молекулярный докинг, prostate cancer, 17,20-lyase activity of CYP17A1, pharmacophore modeling, pharmacophore screening, pharmacokinetic parameters, flexible molecular docking, рак предстательной железы, 17,20-лиазная активность CYP17A1, фармакофорное моделирование, фармакофорный скрининг, фармакокинетические параметры
File Description: application/pdf
Relation: https://doklady.belnauka.by/jour/article/view/1243/1244; Non-steroidal CYP17A1 Inhibitors: Discovery and Assessment / T. Wrobel, F. S. Jorgensen, A. V. Pandey [et al.] // Journal of Medicinal Chemistry. – 2023. – Vol. 66, N 10. – P. 6542–6566. https://doi.org/10.1021/acs.jmedchem.3c00442; Prospective computational design and in vitro bio-analytical tests of new chemical entities as potential selective CYP17A1 lyase inhibitors / N. J. Gumede, W. Nxumalo, K. Bisetty [et al.] // Bioorganic Chemistry. – 2020. – Vol. 94. – Art. 103462. https://doi.org/10.1016/j.bioorg.2019.103462; Рак предстательной железы: лечение и диагностика // Республиканский научно-практический центр онкологии и медицинской радиологии имени Н. Н. Александрова. – URL: https://omr.by/lechenie-opukholej/urologicheskie-opukholi/rak-predstatelnoj-zhelezy (дата обращения: 11.06.2024).; Bird, I. M. The hunt for a selective 17,20 lyase inhibitors: learning lessons from nature / I. M. Bird, D. H. Abbott // Journal of Steroid Biochemistry and Molecular Biology. – 2016. – Vol. 163. – P. 136–146. https://doi.org/10.1016/j.jsbmb.2016.04.021; Promising tools in prostate cancer research: selective non-steroidal cytochrome P450 17A1 inhibitors / S. Bonomo, C. H. Hansen, E. M. Petrunak [et al.] // Scientific Reports. – 2016. – Vol. 6. – Art. 29468. https://doi.org/10.1038/srep29468; Structural and functional evaluation of clinically relevant inhibitors of steroidogenic cytochrome P450 17A1 / E. M. Petrunak, S. A. Rogers, J. Aubé, E. E. Scott // Drug Metabolism and Disposition. – 2017. – Vol. 45, N 6. – P. 635–645. https://doi.org/10.1124/dmd.117.075317; Диченко, Я. В. Компьютерное моделирование строения и реакционной способности молекул / Я. В. Диченко. – Минск, 2023. – 139 с.; Pharmacophore modeling and its applications / R. Tyagi, A. Singh, K. Chaudhary, M. Yadav // Bioinformatics. – 2022. – Vol. 1. – P. 269–289. https://doi.org/10.1016/B978-0-323-89775-4.00009-2; C-(17,20)-lyase inhibitors. Part 2: design, synthesis and structure-activity relationships of (2-naphthylmethyl)-1Himidazoles as novel C-(17,20)-lyase inhibitors / N. Matsunaga, T. Kaku, A. Ojida [et al.] // Bioorganic and Medicinal Chemistry. – 2004. – Vol. 12, N 16. – P. 4313–4336. https://doi.org/10.1016/j.bmc.2004.06.016; 17,20-Lyase inhibitors. Part 4: Design, synthesis and structure-activity relationships of naphthylmethylimidazole derivatives as novel 17,20-lyase inhibitors / T. Kaku, N. Matsunaga, A. Ojida [et al.] // Bioorganic and Medicinal Chemistry. – 2011. – Vol. 19, N 5. – P. 1751–1770. https://doi.org/10.1016/j.bmc.2011.01.017; Pharmacophore model-based virtual screening workflow for discovery of inhibitors targeting Plasmodium falciparum Hsp90 / O. Mafethe, T. Ntseane, T. H. Dongola [et al.] // ACS Omega. – 2023. – Vol. 8, N 41. – P. 38220–38232. https://doi.org/10.1021/acsomega.3c04494; Pharmacophore modeling and 3D QSAR analysis of isothiazolidinedione derivatives as PTP1B inhibitors / G. S. Deora, P. Joshi, V. Rathore [et al.] // Medicinal Chemistry Research. – 2012. – Vol. 22. – P. 3478–3484. https://doi.org/10.1007/s00044-012-0349-7; 3D-QSAR pharmacophore modeling and in silico screening of phospholipase A2α inhibitors / S. V. Jain, M. Ghate, K. Bhadoriya [et al.] // Medicinal Chemistry Research. – 2012. – Vol. 22. – P. 3096–3108. https://doi.org/10.1007/s00044-012-0316-3; ADMETlab 3.0. – URL: https://admetlab3.scbdd.com (date of access: 17.06.2024).; Slow-, tight-binding inhibition of CYP17A1 by abiraterone redefines its kinetic selectivity and dosing regimen / E. Cheong, P. C. Nair, R. W. Y. Neo [et al.] // Journal of Pharmacology and Experimental Therapeutics. – 2020. – Vol. 374, N 3. – P. 438–451. https://doi.org/10.1124/jpet.120.265868; https://doklady.belnauka.by/jour/article/view/1243
-
11Academic Journal
Authors: B. Ya. Alekseev, V. M. Perepukhov, K. M. Nyushko, Б. Я. Алексеев, В. М. Перепухов, К. М. Нюшко
Source: Cancer Urology; Том 19, № 4 (2023); 167-175 ; Онкоурология; Том 19, № 4 (2023); 167-175 ; 1996-1812 ; 1726-9776
Subject Terms: тройная комбинация, nonmetastatic castration-resistant prostate cancer, metastatic hormone-sensitive prostate cancer, high progression risk, combination therapy, double combination, triple combination, неметастатический кастрационно-рефрактерный рак предстательной железы, метастатический гормоночувствительный рак предстательной железы, высокий риск прогрессирования, комбинированная терапия, двойная комбинация
File Description: application/pdf
Relation: https://oncourology.abvpress.ru/oncur/article/view/1790/1503; Шахзадова А.О., Старинский В.В., Лисичникова И.В. Состояние онкологической помощи населению России в 2022 году. Сибирский онкологический журнал 2023;22(5):5–13. DOI:10.21294/1814-4861-2023-22-5-5-13; Tannock I.F., de Wit R., Berry W.R. et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med 2004;351(15):1502–12. DOI:10.1056/NEJMoa040720; Kyriakopoulos C.E., Chen Y.H., Carducci M.A. et al. Chemohormonal therapy in metastatic hormone-sensitive prostate cancer: long-term survival analysis of the randomized phase III E3805 CHAARTED trial. J Clin Oncol 2018;36(11):1080–7. DOI:10.1200/JCO.2017.75.3657; Fizazi K., Tran N., Fein L. et al. Abiraterone plus prednisone in metastatic, castration-sensitive prostate cancer. N Engl J Med 2017;377(4):352–60. DOI:10.1056/NEJMoa1704174; Davis I.D., Martin A.J., Stockler M.R. et al. Enzalutamide with standard first-line therapy in metastatic prostate cancer. N Engl J Med 2019;381(2):121–31. DOI:10.1056/NEJMoa1903835; Armstrong A.J., Szmulewitz R.Z., Petrylak D.P. et al. ARCHES: a randomized, phase III study of androgen deprivation therapy with enzalutamide or placebo in men with metastatic hormonesensitive prostate cancer. J Clin Oncol 2019;37(32):2974–86. DOI:10.1200/JCO.19.00799; Chi K.N., Agarwal N., Bjartell A. et al. Apalutamide for metastatic, castration-sensitive prostate cancer. N Engl J Med 2019;381(1):13–24. DOI:10.1056/NEJMoa1903307; Fizazi K., Foulon S., Carles J. et al. Abiraterone plus prednisone added to androgen deprivation therapy and docetaxel in de novo metastatic castration-sensitive prostate cancer (PEACE-1): a multicentre, open-label, randomised, phase 3 study with a 2×2 factorial design. Lancet 2022;399(10336):1695–707. DOI:10.1016/S0140-6736(22)00367-1; Markham A., Duggan S. Darolutamide: first approval. Drugs 2019;79(16):1813–8. DOI:10.1007/s40265-019-01212-y; Smith M.R., Hussain M., Saad F. et al. Darolutamide and survival in metastatic, hormone-sensitive prostate cancer. N Engl J Med 2022;386(12):1132–42. DOI:10.1056/NEJMoa2119115; Bowling G.C., Dimitrakoff J.D. Darolutamide in metastatic prostate cancer. N Engl J Med 2022;386(24):2344. DOI:10.1056/NEJMc2205310; Sweeney C.J., Chen Y.H., Carducci M. et al. Chemohormonal therapy in metastatic hormone-sensitive prostate cancer. N Engl J Med 2015;373(8):737–46. DOI:10.1056/NEJMoa1503747; Clarke N.W., Ali A., Ingleby F.C. et al. Addition of docetaxel to hormonal therapy in low- and high-burden metastatic hormone sensitive prostate cancer: long-term survival results from the STAMPEDE trial [published correction appears in Ann Oncol 2020;31(3):442]. Ann Oncol 2019;30(12):1992–2003. DOI:10.1093/annonc/mdz396; Fizazi K., Tran N., Fein L. et al. Abiraterone acetate plus prednisone in patients with newly diagnosed high-risk metastatic castration-sensitive prostate cancer (LATITUDE): final overall survival analysis of a randomised, double-blind, phase 3 trial. Lancet Oncol 2019;20(5):686–700. DOI:10.1016/S1470-2045(19)30082-8; James N.D., de Bono J.S., Spears M.R. et al. Abiraterone for prostate cancer not previously treated with hormone therapy. N Engl J Med 2017;377(4):338–51. DOI:10.1056/NEJMoa1702900; Hussain M., Tombal B., Saad F. et al. Darolutamide plus androgendeprivation therapy and docetaxel in metastatic hormone-sensitive prostate cancer by disease volume and risk subgroups in the phase III ARASENS trial. J Clin Oncol 2023;41(20):3595–607. DOI:10.1200/JCO.23.00041; Preisser F., Chun F.K., Banek S. et al. Management and treatment options for patients with de novo and recurrent hormone-sensitive oligometastatic prostate cancer. Prostate Int 2021;9(3):113–8. DOI:10.1016/j.prnil.2020.12.003; Wenzel M., Würnschimmel C., Nocera L. et al. Overall survival after systemic treatment in high-volume versus low-volume metastatic hormone-sensitive prostate cancer: systematic review and network meta-analysis. Eur Urol Focus 2022;8(2):399–408. DOI:10.1016/j.euf.2021.04.003; Mandel P., Hoeh B., Wenzel M. et al. Triplet or doublet therapy in metastatic hormone-sensitive prostate cancer patients: a systematic review and network meta-analysis. Eur Urol Focus 2023;9(1):96–105. DOI:10.1016/j.euf.2022.08.007; Sathianathen N.J., Pan H.Y.C., Lawrentschuk N. et al. Emergence of triplet therapy for metastatic castration-sensitive prostate cancer: an updated systematic review and network meta-analysis. Urol Oncol 2023;41(5):233–9. DOI:10.1016/j.urolonc.2022.10.016; Yanagisawa T., Rajwa P., Thibault C. et al. Androgen receptor signaling inhibitors in addition to docetaxel with androgen deprivation therapy for metastatic hormone-sensitive prostate cancer: a systematic review and meta-analysis. Eur Urol 2022;82(6):584–98. DOI:10.1016/j.eururo.2022.08.002; Maiorano B.A., de Giorgi U., Roviello G. et al. Addition of androgen receptor-targeted agents to androgen-deprivation therapy and docetaxel in metastatic hormone-sensitive prostate cancer: a systematic review and meta-analysis. ESMO Open 2022;7(5):100575. DOI:10.1016/j.esmoop.2022.100575; Roy S., Sayyid R., Saad F. et al. Addition of docetaxel to androgen receptor axis-targeted therapy and androgen deprivation therapy in metastatic hormone-sensitive prostate cancer: a network metaanalysis. Eur Urol Oncol 2022;5(5):494–502. DOI:10.1016/j.euo.2022.06.003; Mori K., Mostafaei H., Sari Motlagh R. et al. Systemic therapies for metastatic hormone-sensitive prostate cancer: network metaanalysis. BJU Int 2022;129(4):423–33. DOI:10.1111/bju.15507; Chi K.N. LATITUDE study – final analysis of phase III study in patients with newly diagnosed high-risk metastatic castrationnaïve prostate cancer. Presented at the American Society of Clinical Oncology Genitourinary Cancers Symposium, February 14–16, 2019, San Francisco, CA.; Chi K.N., Chowdhury S., Bjartell A. et al. Apalutamide in patients with metastatic castration-sensitive prostate cancer: final survival analysis of the randomized, double-blind, phase III TITAN study. J Clin Oncol 2021;39(20):2294–303. DOI:10.1200/JCO.20.03488; Armstrong A. Final overall survival analysis from ARCHES: a phase 3, randomized, double-blind, placebo-controlled study of enzalutamide + ADT in men with mHSPC. Presented at the 2021 European Society for Medical Oncology Annual Congress, September 16–21, 2021.; Gravis G., Boher J.M., Joly F. et al. Androgen deprivation therapy (ADT) plus docetaxel versus ADT alone in metastatic non castrate prostate cancer: impact of metastatic burden and longterm survival analysis of the randomized phase 3 GETUG-AFU15 trial. Eur Urol 2016;70(2):256–62. DOI:10.1016/j.eururo.2015.11.005; James N.D. Abiraterone acetate plus prednisolone for hormonenaïve prostate cancer: long-term results from metastatic (M1) patients in the STAMPEDE randomized trial. Presented at the 2020 European Society for Medical Oncology Virtual Congress, September 19–21, 2020.; Gravis G., Fizazi K., Joly F. et al. Androgen-deprivation therapy alone or with docetaxel in non-castrate metastatic prostate cancer (GETUG-AFU 15): a randomised, open-label, phase 3 trial. Lancet Oncol 2013;14(2):149–58. DOI:10.1016/S1470-2045(12)70560-0; Dou M., Liang H., Liu Y. et al. Based on ARASENS trial: efficacy and safety of darolutamide as an emerging option of endocrinotherapy for metastatic hormone-sensitive prostate cancer-an updated systematic review and network meta-analysis. J Cancer Res Clin Oncol 2023;149(10):7017–27. DOI:10.1007/s00432-023-04658-6; Borgmann H., Lallous N., Ozistanbullu D. et al. Moving towards precision urologic oncology: targeting enzalutamide-resistant prostate cancer and mutated forms of the androgen receptor using the novel inhibitor darolutamide (ODM-201). Eur Urol 2018;73(1):4–8. DOI:10.1016/j.eururo.2017.08.012; Shore N., Zurth C., Fricke R. et al. Evaluation of clinically relevant drug-drug interactions and population pharmacokinetics of darolutamide in patients with nonmetastatic castration-resistant prostate cancer: results of pre-specified and post hoc analyses of the phase III ARAMIS trial. Target Oncol 2019;14(5):527–39. DOI:10.1007/s11523-019-00674-0; Fizazi K., Shore N., Tammela T.L. et al. Darolutamide in nonmetastatic, castration-resistant prostate cancer [published correction appears in N Engl J Med 2022;387(9):860]. N Engl J Med 2019;380(13):1235–46. DOI:10.1056/NEJMoa1815671; https://oncourology.abvpress.ru/oncur/article/view/1790
-
12Academic Journal
Authors: Volgareva G.M.
Contributors: The work was performed without external funding, Работа выполнена без спонсорской поддержки
Source: Advances in Molecular Oncology; Vol 11, No 2 (2024); 29-39 ; Успехи молекулярной онкологии; Vol 11, No 2 (2024); 29-39 ; 2413-3787 ; 2313-805X
Subject Terms: oncogenic papillomaviruses, breast cancer, prostate cancer, lung cancer, colorectal cancer, онкогенные папилломавирусы, рак молочной железы, рак предстательной железы, рак легкого, рак толстой и прямой кишок
File Description: application/pdf
Relation: https://umo.abvpress.ru/jour/article/view/674/345; https://umo.abvpress.ru/jour/article/view/674
-
13Academic Journal
Authors: Shutko E.V., Bryzgunova O.E., Ostal’cev I.A., Pak S.V., Krasi’nikov S.E., Laktionov P.P., Konoshenko M.Y.
Contributors: The research was carried out at the expense of a grant from the Russian Science Foundation (grant No. 23-25-10026, https://rscf.ru/project/23-25-10026) within the framework of the project 0000005406995998235120582 supported by the Government of the Novosibirsk Region No. r-45, Исследование выполнено за счет гранта Российского научного фонда (грант № 23-25-10026, https://rscf.ru/project/23-25-10026) в рамках поддержанного Правительством Новосибирской области проекта 0000005406995998235120582 № р-45
Source: Advances in Molecular Oncology; Vol 11, No 1 (2024); 55-78 ; Успехи молекулярной онкологии; Vol 11, No 1 (2024); 55-78 ; 2413-3787 ; 2313-805X
Subject Terms: prostate cancer, miRNA, extracellular vesicles of urine, radical prostatectomy, reverse transcription polymerase chain reaction, miRNA dynamics after cancer treatment, рак предстательной железы, микроРНК, внеклеточные везикулы мочи, радикальная простатэктомия, полимеразная цепная реакция с обратной транскрипцией, динамика микроРНК после лечения рака
File Description: application/pdf
Relation: https://umo.abvpress.ru/jour/article/view/649/338; https://umo.abvpress.ru/jour/article/view/649
-
14Academic Journal
Authors: T. P. Berezovskaya, V. O. Ripp, A. V. Troyanov, S. A. Ivanov, A. D. Kaprin, Т. П. Березовская, В. О. Рипп, А. В. Троянов, С. А. Иванов, А. Д. Каприн
Source: Diagnostic radiology and radiotherapy; Том 15, № 3 (2024); 19-31 ; Лучевая диагностика и терапия; Том 15, № 3 (2024); 19-31 ; 2079-5343
Subject Terms: Met-RADS-P, diffusion-weighted whole-body imaging, metastatic prostate cancer, диффузионно-взвешенная томография всего тела, метастатический рак предстательной железы
File Description: application/pdf
Relation: https://radiag.bmoc-spb.ru/jour/article/view/1018/654; Padhani A.R., Liu G., Koh D.M. et al. Diffusion-weighted magnetic resonance imaging as a cancer biomarker: consensus and recommendations // Neoplasia. 2009. Vol. 11, No. 2. Р. 102–125. doi:10.1593/neo.81328.; Summers P. Saia G., Colombo A. et al. Whole-body magnetic resonance imaging: technique, guidelines and key applications // Ecancer medical science. 2021. Vol. 15. Р. 1164. doi:10.3332/ecancer.2021.1164.; Messiou C., Hillengass J., Delorme S. et al. Guidelines for acquisition, interpretation, and reporting of whole-body MRI in myeloma: myeloma response assessment and diagnosis system (MY-RADS) // Radiology. 2019. Vol. 291. Р. 5–13. doi:10.1148/radiol.2019181949.; Petralia G., Koh D.M., Attariwala R. et al. Oncologically Relevant Findings Reporting and Data System (ONCO-RADS): Guidelines for the Acquisition, Interpretation, and Reporting of Whole-Body MRI for Cancer Screening // Radiology. 2021. Vol. 299, No. 3. Р. 494–507. doi:10.1148/radiol.2021201740.; Padhani A.R., Lecouvet F.E., Tunariu N. et al. METastasis Reporting and Data System for Prostate Cancer: Practical Guidelines for Acquisition, Interpretation, and Reporting of Whole-body Magnetic Resonance Imaging-based Evaluations of Multiorgan Involvement in Advanced Prostate Cancer // Eur. Urol. 2017. Vol. 71, No. 1. Р. 81–92. doi:10.1016/j.eururo.2016.05.033.; Schmidt G.P., Schoenberg S.O., Reiser M.F. et al. Whole-body MR imaging of bone marrow // Eur. J. Radiol. 2005. Vol. 55. Р. 33–40. doi:10.1016/j.ejrad.2005.01.019.; Dixon WT. Simple proton spectroscopic imaging // Radiology. 1984. Vol. 153, No. 1. Р. 189–194. doi:10.1148/radiology.153.1.6089263.; Bray T.J., Chouhan M.D., Punwani S., Bainbridge A., Hall-Craggs M.A. Fat fraction mapping using magnetic resonance imaging: insight into pathophysiology // Br. J. Radiol. 2018 Sep. Vol. 91, No. 1089. Р. 20170344. doi:10.1259/bjr.20170344.; Gee C.S., Nguyen J.T., Marquez C.J., Heunis J., Lai A., Wyatt C., Han M., Kazakia G., Burghardt A.J., Karampinos D.C., Carballido-Gamio J., Krug R. Validation of bone marrow fat quantification in the presence of trabecular bone using MRI // J. Magn. Reson. Imaging. 2015. Vol. 42, No. 2. Р. 539–544. doi:10.1002/jmri.24795.; Guerini H., Omoumi P., Guichoux F. et al. Fat suppression with Dixon techniques in musculoskeletal magnetic resonance imaging: a pictorial review // Semin Musculoskeletal Radiol. 2015. Vol. 19. Р. 335–347.; Lins C.F., Salmon C.E.G., Nogueira-Barbosa M.H. Applications of the Dixon technique in the evaluation of the musculoskeletal system // Radiol. Bras. 2021. Vol. 54, No. 1. Р. 33–42. doi:10.1590/0100-3984.2019.0086.; Lecouvet F.E., Pasoglou V., Van Nieuwenhove S., Van Haver T., de Broqueville Q., Denolin V. et al. Shortening the acquisition time of whole-body MRI: 3D T1 gradient echo Dixon vs fast spin echo for metastatic screening in prostate cancer // Eur. Radiol. 2020. Vol. 30, No. 6. Р. 3083–3093.; Михайлов А.И., Панов В.О., Мещерякова Н.А., Пронин А.И., Вихрова Н.Б., Долгушин М.Б. Диффузионно-взвешенная магнитно-резонансная томография всего тела с оценкой измеряемого коэффициента диффузии при лимфоме Ходжкина // Онкологический журнал: лучевая диагностика, лучевая терапия. 2018. Vol. 1, № 3. С. 44–48. doi:10.37174/2587-7593-2018-1-3-44-48; Kwee T.C., Takahara T., Ochiai R., Nievelstein R.A., Luijten P.R. Diffusion-weighted whole-body imaging with background body signal suppression (DWIBS): features and potential applications in oncology // Eur. Radiol. 2008. Vol. 18, No. 9. Р. 1937–1952. doi:10.1007/s00330-008-0968-z.; Takahara et al. Diffusion weighted whole body imaging with background body signal suppression (DWIBS): Technical improvement using free breathing, STIR and high resolution 3D display // Radiation medicine. 2003. Vol. 22. Р. 275–282.; Nagarajan R., Margolis D., Raman S., Sheng К., King Ch., Reiter R., Thomas M.A. Correlation of Gleason Scores with Diffusion-Weighted Imaging Findings of Prostate Cancer // Advances in Urology. 2012. Vol. 2012, 5 p. https://doi.org/10.1155/2012/374805.; Manetta R., Palumbo P., Gianneramo C. et al. Correlation between ADC values and Gleason score in evaluation of prostate cancer: multicentre experience and review of the literature // Gland Surg. 2019. Vol. 8, Suppl. 3. Р. S216-S222. doi:10.21037/gs.2019.05.02.; Shi Y.J., Li X.T., Zhang X.Y., Liu Y.L., Tang L., Sun Y.S. Differential diagnosis of hemangiomas from spinal osteolytic metastases using 3.0 T MRI: comparison of T1-weighted imaging, chemical-shift imaging, diffusion-weighted and contrast-enhanced imaging // Oncotarget. 2017. Vol. 8, No. 41. Р. 71095–71104. doi:10.18632/oncotarget.20533.; Koh D.M., Blackledge M., Padhani A.R. et al. Whole-body diffusion-weighted MRI: tips, tricks, and pitfalls // AJR Am. J. Roentgenol. 2012. Vol. 199, No. 2. Р. 252– 262. doi:10.2214/AJR.11.7866.; Van Nieuwenhove S., Van Damme J., Padhani A.R., Vandecaveye V., Tombal B., Wuts J., Pasoglou V., Lecouvet F.E. Whole-body magnetic resonance imaging for prostate cancer assessment: Current status and future directions // J. Magn. Reson. Imaging. 2022. 55: 653–80. https://doi.org/10.1002/jmri.27485.; Grimm A., Meyer H., Nickel M.D. et al. Evaluation of 2-point, 3-point, and 6-point Dixon magnetic resonance imaging with flexible echo timing for muscle fat quantification // Eur. J. Radiol. 2018. Vol. 103. Р. 57–64.; Gandaglia G., Abdollah F., Schiffmann J., Trudeau V., Shariat SF., Kim SP., Perrotte P., Montorsi F., Briganti A., Trinh QD., Karakiewicz PI., Sun M. Distribution of metastatic sites in patients with prostate cancer: A population-based analysis // Prostate. 2014. Feb. Vol. 74, No. 2. Р. 210–216. doi:10.1002/pros.22742.; Scher H.I., Morris M.J., Stadler W.M. et al. Trial Design and Objectives for Castration-Resistant Prostate Cancer: Updated Recommendations From the Prostate Cancer Clinical Trials Working Group 3 // J. Clin. Oncol. 2016. Vol. 34, No. 12. Р. 1402–1418. doi:10.1200/JCO.2015.64.2702.; Lapini A., Caffo O., Pappagallo G., Iacovelli R., D’Angelillo R.M., Vavassori V., Ceccarelli R., Bracarda S., Jereczek-Fossa B.A., Da Pozzo L., Conti G.N. Monitoring Patients with Metastatic Hormone-Sensitive and Metastatic Castration-Resistant Prostate Cancer: A Multidisciplinary Consensus Document // Cancers (Basel). 2019 Dec 1. Vol. 11, No. 12. Р. 1908. doi:10.3390/cancers11121908.; Eisenhauer E.A., Therasse P., Bogaerts J. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) // Eur. J. Cancer. 2009. Vol. 45. Р. 228–247.; Pricolo P., Ancona E., Summers P. et al. Whole-body magnetic resonance imaging (WB-MRI) reporting with the METastasis Reporting and Data System for Prostate Cancer (MET-RADS-P): inter-observer agreement between readers of different expertise levels // Cancer Imaging. 2020. Vol. 20, No. 1. Р. 77. doi:10.1186/s40644-020-00350-x.; Liu X., Xie T., Sun Z., Guo Y., Zhang X., Wang X. Analysis of interreader agreement in structured reports of pelvic multiparametric magnetic resonance imaging using the METastasis Reporting and Data System for Prostate Cancer guidelines // Diagn. Interv. Radiol. 2023. Jan 31. Vol. 29, No. 1. Р. 29–39. doi:10.5152/dir.2022.211232.; Yoshida S., Takahara T., Ishii C. et al. METastasis Reporting and Data System for Prostate Cancer as a Prognostic Imaging Marker in Castration-resistant Prostate Cancer // Clin. Genitourin Cancer. 2020. Vol. 18, No. 4. Р. e391-e396. doi:10.1016/j.clgc.2019.12.010.
-
15Academic Journal
Authors: M. D. Ter-Ovanesov, D. M. Yagudaev, A. A. Gritskevich, E. V. Anikanova, K. I. Medvedev, М. Д. Тер-Ованесов, Д. М. Ягудаев, А. А. Грицкевич, Е. В. Аниканова, К. И. Медведев
Contributors: this work was not funded., финансирование данной работы не проводилось.
Source: Research and Practical Medicine Journal; Том 11, № 2 (2024); 89-100 ; Research'n Practical Medicine Journal; Том 11, № 2 (2024); 89-100 ; 2410-1893 ; 10.17709/2410-1893-2024-11-2
Subject Terms: рак желудка, primary multiple synchronous cancer, primary multiple metachronous cancer, urothelial cancer, prostate cancer, renal cell carcinoma, gastric cancer, clinical case, синхронный рак, метахронный рак, уротелиальный рак, рак предстательной железы, рак почки
File Description: application/pdf
Relation: https://www.rpmj.ru/rpmj/article/view/917/617; https://www.rpmj.ru/rpmj/article/view/917/631; Злокачественные новообразования в России в 2021 году (заболеваемость и смертность). Под ред. Каприна А. Д., Старинского В. В., Шахзадовой А. О. М.: МНИОИ им. П. А. Герцена – филиал ФГБУ «НМИЦ радиологии» Минздрава России, 2022, 252 с. Доступно по: https://oncology-association.ru/wp-content/uploads/2022/11/zlokachestvennye-novoobrazovaniya-vrossii-v-2021-g_zabolevaemost-i-smertnost.pdf; Billroth T. Handbuch fur Studierende and Artze, 14. Die allgemeine chirurgische pathologie und therapie. Berlin: Auflage; 1889.; Warren S, Gates O. Multiple primary malignant tumors: a survey of the literature and a statistic study. Am J Cancer. 1932;16:1358–1414.; Moertel CG. Multiple primary malignant neoplasms: historical perspectives. Cancer. 1977;40:1786–1792. https://doi.org/10.1002/1097-0142(197710)40:4+3.0.co;2-2; Сельчук В. Ю. Первично-множественные злокачественные опухоли (клиника, лечение и закономерности развития). Дисс. … докт. мед. наук. М., 1994.; Lv M, Zhang X, Shen Y, Wang F, Yang J, Wang B, et al. Clinical analysis and prognosis of synchronous and metachronous multiple primary malignant tumors. Medicine (Baltimore). 2017 Apr;96(17):e6799. https://doi.org/10.1097/md.0000000000006799; Liu Z, Jin C, Zhang Y, Jiang Y, Wang J, Zheng L. Identification of BRAF, CCND1, and MYC mutations in a patient with multiple primary malignant tumors: a case report and review of the literature. World J Surg Oncol. 2023 May 24;21(1):158. https://doi.org/10.1186/s12957-023-03036-3; Zheng R, Li H, Ye Y, Guan L, Li H, Ye Z, et al. Clinicopathological features and prognostic analysis of 77 patients with multiple primary cancers. J BUON. 2020 Jul-Aug;25(4):2110–2116.; Muller J, Grosclaude P, Lapôtre-Ledoux B, Woronoff AS, Guizard AV, Bara S, et al. Trends in the risk of second primary cancer among bladder cancer survivors: a population-based cohort of 10 047 patients. BJU Int. 2016 Jul;118(1):53–59. https://doi.org/10.1111/bju.13351; Гладков О. А., Зуков Р. А., Матвеев В. Б., Митин Т., Носов Д. А., Попов А. М. Практические рекомендации по лекарственному лечению рака мочевого пузыря. Практические рекомендации RUSSCO. Злокачественные опухоли. 2022;12(3S2):589–606. https://doi.org/10.18027/2224-5057-2022-12-3s2-589-606; Sylvester RJ, Oosterlinck W, Holmang S, Sydes MR, Birtle A, Gudjonsson S, et al. Systematic Review and Individual Patient Data Meta-analysis of Randomized Trials Comparing a Single Immediate Instillation of Chemotherapy After Transurethral Resection with Transurethral Resection Alone in Patients with Stage pTa-pT1 Urothelial Carcinoma of the Bladder: Which Patients Benefit from the Instillation? Eur Urol. 2016 Feb;69(2):231–244. https://doi.org/10.1016/j.eururo.2015.05.050; Носов Д. А., Волкова М. И., Гладков О. А., Карабина Е. В., Крылов В. В., Матвеев В. Б., и др. Практические рекомендации по лекарственному лечению рака предстательной железы. Практические рекомендации RUSSCO, часть 1. Злокачественные опухоли. 2023;13(3S2-1):640–660. https://doi.org/10.18027/2224-5057-2023-13-3s2-1-640-660; The compilation of the complete Guidelines should be referenced as: EAU Guidelines. Edn. presented at the EAU Annual Congress Milan 2023. ISBN 978-94-92671-19-6.; Bratan F, Niaf E, Melodelima C, Chesnais AL, Souchon R, Mège-Lechevallier F, et al. Influence of imaging and histological factors on prostate cancer detection and localisation on multiparametric MRI: a prospective study. Eur Radiol. 2013 Jul;23(7):2019–2029. https://doi.org/10.1007/s00330-013-2795-0; Borofsky S, George AK, Gaur S, Bernardo M, Greer MD, Mertan FV, et al. What Are We Missing? False-Negative Cancers at Multiparametric MR Imaging of the Prostate. Radiology. 2018 Jan;286(1):186–195. https://doi.org/10.1148/radiol.2017152877; Dickinson L, Ahmed HU, Allen C, Barentsz JO, Carey B, Futterer JJ, et al. Magnetic resonance imaging for the detection, localisation, and characterisation of prostate cancer: recommendations from a European consensus meeting. Eur Urol. 2011 Apr;59(4):477– 494. https://doi.org/10.1016/j.eururo.2010.12.009; Turkbey B, Rosenkrantz AB, Haider MA, Padhani AR, Villeirs G, Macura KJ, et al. Prostate Imaging Reporting and Data System Version 2.1: 2019 Update of Prostate Imaging Reporting and Data System Version 2. Eur Urol. 2019 Sep;76(3):340–351. https://doi.org/10.1016/j.eururo.2019.02.033; Nishiyama N, Hotta H, Takahashi A, Yanase M, Itoh N, Tachiki H, et al. Upper tract urothelial carcinoma following intravesical bacillus Calmette-Guérin therapy for nonmuscle-invasive bladder cancer: Results from a multi-institutional retrospective study. Urol Oncol. 2018 Jun;36(6):306.e9-306.e15. https://doi.org/10.1016/j.urolonc.2018.02.009; Cowan NC, Turney BW, Taylor NJ, McCarthy CL, Crew JP. Multidetector computed tomography urography for diagnosing upper urinary tract urothelial tumour. BJU Int. 2007 Jun;99(6):1363–1370. https://doi.org/10.1111/j.1464-410x.2007.06766.x; Janisch F, Shariat SF, Baltzer P, Fajkovic H, Kimura S, Iwata T, et al. Diagnostic performance of multidetector computed tomographic (MDCTU) in upper tract urothelial carcinoma (UTUC): a systematic review and meta-analysis. World J Urol. 2020 May;38(5):1165– 1175. https://doi.org/10.1007/s00345-019-02875-8; Sharma V, Miest TS, Juvet TS, Toussi A, Packiam V, Chamie K, et al. The Impact of Upper Tract Urothelial Carcinoma Diagnostic Modality on Intravesical Recurrence after Radical Nephroureterectomy: A Single Institution Series and Updated Meta-Analysis. J Urol. 2021 Sep;206(3):558–567. https://doi.org/10.1097/ju.0000000000001834; Ayyathurai R, Soloway MS. Monitoring of the upper urinary tract in patients with bladder cancer. Indian J Urol. 2011 Apr;27(2):238– 244. https://doi.org/10.4103/0970-1591.82844; Fritsche HM, Burger M, Svatek RS, Jeldres C, Karakiewicz PI, Novara G, et al. Characteristics and outcomes of patients with clinical T1 grade 3 urothelial carcinoma treated with radical cystectomy: results from an international cohort. Eur Urol. 2010 Feb;57(2):300–309. https://doi.org/10.1016/j.eururo.2009.09.024; Turker P, Bostrom PJ, Wroclawski ML, van Rhijn B, Kortekangas H, Kuk C, et al. Upstaging of urothelial cancer at the time of radical cystectomy: factors associated with upstaging and its effect on outcome. BJU Int. 2012 Sep;110(6):804–811. https://doi.org/10.1111/j.1464-410x.2012.10939.x; Moschini M, Sharma V, Dell'oglio P, Cucchiara V, Gandaglia G, Cantiello F, et al. Comparing long-term outcomes of primary and progressive carcinoma invading bladder muscle after radical cystectomy. BJU Int. 2016 Apr;117(4):604–610. https://doi.org/10.1111/bju.13146; https://www.rpmj.ru/rpmj/article/view/917
Availability: https://www.rpmj.ru/rpmj/article/view/917
-
16Academic Journal
Authors: D. G. Zaridze, Д. Г. Заридзе
Source: Public Health; Том 4, № 4 (2024); 24-42 ; Общественное здоровье; Том 4, № 4 (2024); 24-42 ; 2949-1274 ; 2782-1676
Subject Terms: рак шейки матки, artificial intelligence, AI, cancer of the breast, lung, prostate, large bowel, cervix, искусственный интеллект, ИИ, рак молочной железы, рак предстательной железы, рак легкого, рак толстой кишки
File Description: application/pdf
Relation: https://ph.elpub.ru/jour/article/view/229/129; Breast Cancer Screening. IARC handbook of Cancer Prevention, International Agency for Research on Cancer, WHO, IARC Pres. 2002.; European Commission, European Guidelines for Quality Assurance in Breast Cancer Screening and Diagnosis, 2006.; Cervix cancer screening. IARC Handbooks of cancer Prevention, International Agency for Research on Cancer, World Health Organization, IARC Press, 2005.; European Commission. European Guidelines for Quality Assurance in Cervical Cancer Screening, 2nd ed. – Luxembourg: Office for Official Publications of the European Communities, 2008.; Заридзе Д. Г. Профилактика рака. Руководство для врачей. – М.: ИМА-ПРЕСС, 2009. – 224 с. – 9 ил. – 63 табл.; Давыдов М.И., Заридзе Д. Г. Скрининг злокачественных опухолей: современное состояние и перспективы. Вестник Московского Онкологического Общества. 2014; 3(606):2–6.; Барчук А.А., Раскина Ю. В., Смирнова О. В., Беляев А. М., Багненко С. Ф. Скрининг онкологических заболеваний на уровне государственных программ: обзор, рекомендации и управление. Общественное здоровье. 2021; 1(1):19–31.; Zaridze D.G., Boyle P., Smans M. International trends in prostаtic cancer. Int J Cancer. 1984; 33(2): 223–230.; Black W.C., Haggstrom D. A., Welch H. G. All-cause mortality in randomized trials of cancer screening. J Natl Cancer Inst. 2002; 94(3):167–173.; Заридзе Д.Г., Максимович Д. М., Стилиди И. С. Новая парадигма скрининга и ранней диагностики: оценка пользы и вреда. Вопросы онкологии. 2020; 66(6):589–602.; Заридзе Д. Г. Искусственный интеллект повышает эффективность скрининга и диагностики злокачественных опухолей. Национальная онкологическая программа (2030). 2024; 2:32–34.; Tabár L., Vitak B., Chen H. H. et al. Beyond randomized controlled trials: organized mammographic screening substantially reduces breast carcinoma mortality. Cancer. 2001; 91(9):1724–1731.; Tabár L., Yen M. F., Vitak B. et al. Mammography service screening and mortality in breast cancer patients: 20-year follow-up before and after introduction of screening. Lancet. 2003; 361(9367):1405–1410.; Nelson H.D., Tyne K., Naik A. et al. Screening for Breast Cancer: Systematic Evidence Review Update for the US Preventive Services Task Force. Report 10–05142-EF-1. U. S. Preventive Services Task Force Evidence Syntheses, formerly Systematic Evidence Reviews. Rockville, MD: Agency for Health Сare Research and Quality (US). 2009.; Swedish Organised Service Screening Evaluation Group. Reduction in breast cancer mortality from organized service screening with mammography: 1. Further confirmation with extended data. Cancer Epidemiol Biomarkers Prev. 2006; 15(1):45–51.; Pisano E.D., Gatsonis C., Hendrick E. et al. Diagnostic performance of digital versus film mammography for breast-cancer screening. N Engl J Med. 2005; 353(17): 1773–1783.; Eastern Cooperative Oncology Group – American Col lege of Radiology Imaging Network Cancer Research Group. Digital Tomosynthesis Mammography and Digital Mammography in Screening Patients for Breast Cancer (NCT03233191). Philadelphia, PA: Eastern Cooperative Oncology Group – American College of Radiology Imaging Network Cancer Research Group; 2017. Clinicaltrials.gov/ct2/show/NCT03233191. Accessed December 18, 2017.; Majid A.S., de Paredes E. S., Doherty R. D., Sharma N. R., Salvador X. Missed breast carcinoma: pitfalls and pearls. Radiographics. 2003; 23(4):881–895.; Miglioretti D.L., Gard C. C., Carney P. A. et al. When radiologists perform best: the learning curve in screening mammogram interpretation. Radiology. 2009; 253(3): 632–640.; Independent UK Panel on Breast cancer Screening. The benefits and harms of breast cancer screening: an independent review. Lancet. 2012; 380(9855):1778–1786.; Lehman C.D., Arao R. F., Sprague B. L. et al. National Performance Benchmarks for Modern Screening Digital Mammography: Update from the Breast Cancer Surveillance Consortium. Radiology. 2017; 283(1):49–58.; Salim M., Dembrower K., Eklund M., Lindholm P., Strand F. Range of Radiologist Performance in a Population-based Screening Cohort of 1 Million Digital Mammography Examinations. Radiology. 2020; 297(1):33–39.; Kwee T.C., Kwee R. M. Workload of diagnostic radiologists in the foreseeable future based on recent scientific advances: growth expectations and role of artificial intelligence. Insights Imaging. 2021; 12(1):88.; Fenton J.J., Abraham L., Taplin S. H. et al. Effectiveness of computer-aided detection in community mammography practice. J Natl Cancer Inst. 2011; 103(15):1152–1161.; Lehman C.D., Wellman R. D., Buist D. S. et al. Diagnostic Accuracy of Digital Screening Mammography With and Without Computer-Aided Detection. JAMA Intern Med. 2015; 175(11):1828–1837.; Ernster V.L., Ballard-Barbash R., Barlow W. E. et al. Detection of ductal carcinoma in situ in women undergoing screening mammography. J Natl Cancer Inst. 2002; 94(20):1546–1554.; Kim H.-E., Kim H. H., Han B.-K. et al. Changes in cancer detection and false-positive recall in mammography using artificial intelligence: a retrospective, multireader study. Lancet Digit Health. 2020; 2(3): e138-e148.; Dembrower K., Crippa A., Colón E., Eklund M., Strand F.; ScreenTrustCAD Trial Consortium. Artificial intelligence for breast cancer detection in screening mammography in Sweden: a prospective, population-based, paired-reader, non-inferiority study. Lancet Digit Health. 2023; 5(10): e703-e711.; National Lung Screening Trial Research Team; Aberle D. R., Adams A. M., Berg C. D. et al. Reduced lung-cancer mortality with low-dose computed tomographic screening. N Engl J Med. 2011; 365(5):395–409.; De Koning H. J., van der Aalst C. M., de Jong P. A. et al. Reduced Lung-Cancer Mortality with Volume CT Screening in a Randomized Trial. N Engl J Med. 2020; 382(6):503– 513.; Bach P.B., Mirkin J. N., Oliver T. K. et al. Benefits and harms of CT screening for lung cancer: a systematic review. JAMA. 2012; 307(22):2418–2429.; Bonney A., Malouf R., Marchal C. et al. Impact of low‐dose computed tomography (LDCT) screening on lung cancer‐related mortality. Cochrane Database Syst Rev. 2022; 8(8): CD013829.; Ardila D., Kiraly A. P., Bharadwaj S. et al. End-to-end lung cancer screening with three-dimensional deep learning on low-dose chest computed tomography. Nat Med. 2019; 25(6):954–961.; Mikhael PG, Wohlwend J., Yala A. et al. Sybil: A Validated Deep Learning Model to Predict Future Lung Cancer Risk From a Single Low-Dose Chest Computed Tomography. J Clin Oncol. 2023; 41(12):2191–2200.; Andriole G.L., Crawford E. D., Grubb R. L. 3rd et al. Prostate cancer screening in the randomized Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial: mortality results after 13 years of follow-up. J Natl Cancer Inst. 2012; 104(2):125–132.; Schröder F.H., Hugosson J., Roobol M. J. et al. Prostate-cancer mortality at 11 years of follow-up. N Engl J Med. 2012; 366(11):981–990.; Moyer V.A.; U. S. Preventive Services Task Force. Screening for Prostate Cancer: US Preventive Services Task Force Recommendation Statement. Ann Intern Med. 2012; 157(2):120–134.; Carter H.B., Albertsen P. C., Barry M. J. et al. Early Detection of Prostate Cancer: AUA Guideline. J Urol. 2013; 190(2):419–426.; Qaseem A., Barry M. J., Denberg T. D. et al. Screening for prostate cancer: a guidance statement from the Clinical Guidelines Committee of the American College of Physicians. Ann Intern Med. 2013; 158(10):761–769.; Yakar D., Debats O. A., Bomers J. G. et al. Predictive value of MRI in the localization, staging, volume estimation, assessment of aggressiveness, and guidance of radiotherapy and biopsies in prostate cancer. J Magn Reson Imaging. 2012; 35(1):20–31.; Moradi M., Salcudean S. E., Chang S. D. et al. Multiparametric MRI maps for detection and grading of dominant prostate tumors. J Magn Reson Imaging. 2012; 35(6):1403– 1413.; Zhao C., Gao G., Fang D. et al. The efficiency of multiparametric magnetic resonance imaging (mpMRI) using PIRADS Version 2 in the diagnosis of clinically significant prostate cancer. Clin Imaging. 2016; 40(5):885–888.; Kasel-Seibert M., Lehmann T., Aschenbach R. et al. Assessment of PI-RADS v2 for the Detection of Prostate Cancer. Eur J Radiol. 2016; 85(4):726–731.; Fehr D., Veeraraghavan H., Wibmer A. et al. Automatic classification of prostate cancer Gleason scores from multiparametric magnetic resonance images. Proc Natl Acad Sci U S A. 2015; 112(46):6265–6273.; Chen T., Li M., Gu Y. et al. Prostate Cancer Differentiation and Aggressiveness: Assessment With a Radiomic-Based Model vs. PI-RADS v2. J Magn Reson Imaging. 2018; 49(3):875–884.; Zhang H., Ji J., Liu Z. et al. Artificial intelligence for the diagnosis of clinically significant prostate cancer based on multimodal data: a multicenter study. BMC Med. 2023; 21(1):270.; Cuocolo R., Cipullo M. B., Stanzione A. et al. Machine learning for the identification of clinically significant prostate cancer on MRI: a meta-analysis. Eur Radiol. 2020; 30(12):6877–6887.; Clark J.C., Collan Y., Zaridze D. G. et al. Prevalence of polyps in an autopsy series from areas with varying incidence of large-bowel cancer. Int J Cancer. 1985; 36(2):179–186.; Mandel J.S., Church T. R., Bond J. H. et al. The effect of fecal occult-blood screening on the incidence of colorectal cancer. N Engl J Med. 2000; 343(22):1603–1607.; Atkin W.S., Edwards R., Kralj-Hans I. et al.; UK Flexible Sigmoidoscopy Trial Investigators. Once-only flexible sigmoidoscopy screening in prevention of colorectal cancer: a multicentre randomised controlled trial. Lancet. 2010; 375(9726):1624–1633.; Schoen R.E., Pinsky P. F., Weissfeld J. L. et al.; PLCO Proj ect Team. Colorectal-cancer incidence and mortality with screening flexible sigmoidoscopy. N Engl J Med. 2012; 366(25):2345–2357.; Zhu C., Bassig B. A., Zaridze D. et al. A birth cohort analysis of the incidence of ascending and descending colon cancer in the United States, 1973–2008. Cancer Causes Control. 2013; 24(6):1147–1156.; Bretthauer M., Løberg M., Wieszczy P. et al.; NordICC Study Group. Effect of Colonoscopy Screening on Risks of Colorectal Cancer and Related Death. N Engl J Med. 2022; 387(17):1547–1556.; Hassan C., Spadaccini M., Mori Y. et al. Real-Time Computer-Aided Detection of Colorectal Neoplasia During Colonoscopy: A Systematic Review and Meta-analysis. Ann Intern Med. 2023; 176(9):1209–1220.; Lou S., Du F., Song W. et al. Artificial intelligence for colorectal neoplasia detection during colonoscopy: a systematic review and meta-analysis of randomized clinical trials. EClinicalMedicine. 2023; 66:102341.; Maas M.H.J., Neumann H., Shirin H. et al. A computer-aided polyp detection system in screening and surveillance colonoscopy: an international, multicentre, randomised, tandem trial. Lancet Digit Health. 2024; 6(3): е157-e165.; International Agency for Research on Cancer, World Health Organization. Cervix cancer screening. IARC Handbooks of cancer Prevention. IARC. 2005.; Cox J.T., Castle P. E., Behrens C. M. et al.; Athena HPV study group. Comparison of cervical cancer screening strategies incorporating different combinations of cytology, HPV testing, and genotyping for HPV 16/18: results from the ATHENA HPV study. Am J Obstet Gynecol. 2013; 208(3):184.e1–184.e11.; WHO guideline for screening and treatment of cervical pre-cancer lesions for cervical cancer prevention, second edition. Geneva: World Health Organization. 2021.; Заридзе Д.Г., Стилиди И. С., Мукерия А. Ф. Научное обоснование эффективности первичной и вторичной (скрининга) профилактики рака шейки матки. Общественное здоровье. 2022; 2(4):15–23.; Wang J., Yu Y., Tan Y. et al. Artificial intelligence enables precision diagnosis of cervical cytology grades and cervical cancer. Nat Commun. 2024; 15(1);4369.; Солодкий В.А., Каприн А. Д., Нуднов Н. В., Харчен ко Н. В., Запиров Г. М., Дибирова Ш. М., Подоль ская М. В., Кунда М. А. Возможности искусственного интеллекта в оценке риска рака молочной железы на маммографических изображениях (клинические примеры). Вестник российского научного центра рентгенорадиологии (вестник РНЦРР), 2023; 2023(1):25–32.; Солодкий В.А., Каприн А. Д., Нуднов Н. В., Харчен ко Н. В., Ходорович О. С., Запиров Г. М., Шерстнёва Т. В., Дибирова Ш. М., Канахина Л. Б. Современные системы поддержки принятия врачебных решений на базе искусственного интеллекта для анализа цифровых маммографических изображений. Вестник рентгенологии и радиологии. 2023; 104(2):151–162.; Арзамасов К.М., Васильев Ю. А., Владзимирский А. В., Омелянская О. В., Бобровская Т. М., Семенов С. С., Четвериков С. Ф., Кирпичев Ю. С., Павлов Н. А., Ан дрейченко А. Е. Применение компьютерного зрения для профилактических исследований на примере маммографии. Профилактическая медицина. 2023; 26(6):117–123.; Павлова В.И., Белая Ю. А., Воронцов А. Ю., Прище пов А. А., Князев С. М., Михайлов А. А., Ковалева А. В., Аревшатян Э. Г., Палтуев Р. М., Чёрная А. В., Захаро ва Н. А. Результаты научно-исследовательской работы Российского общества онкомаммологов «Использование искусственного интеллекта для раннего выявления рака молочной железы». Опухоли женской репродуктивной системы. 2023; 19(2):54–60.; Arzamasov K., Vasilev Y., Zelenova M. et al. Independent evaluation of the accuracy of 5 artificial intelligence software for detecting lung nodules on chest X-rays. Quant Imaging Med Surg. 2024; 14(8):5288–5303.; Васильев Ю.А., Арзамасов К. М., Колсанов А. В., Владзи мирский А. В., Омелянская О. В., Пестренин Л. Д., Неча ев Н. Б. Опыт применения программного обеспечения на основе технологий искусственного интеллекта на данных 800 тысяч флюорографических исследований. Врач и информационные технологии. 2023; 4:54–65.; Пилюс П.С., Дрокин И. С., Баженова Д. А., Маков ская Л. А., Синицын В. Е. Оценка перспектив использования технологий искусственного интеллекта для анализа КТ-изображений органов грудной клетки с целью выявления признаков злокачественных новообразований в легких. Медицинская визуализация. 2023; 27(2):138–146.; https://ph.elpub.ru/jour/article/view/229
-
17Academic Journal
Authors: V. A. Solodkiy, A. Yu. Pavlov, A. G. Dzidzaria, A. D. Tsibulskii, В. А. Солодкий, А. Ю. Павлов, А. Г. Дзидзария, А. Д. Цыбульский
Source: Cancer Urology; Том 19, № 4 (2023); 86-96 ; Онкоурология; Том 19, № 4 (2023); 86-96 ; 1996-1812 ; 1726-9776
Subject Terms: радикальная простатэктомия, prostate cancer, salvage brachytherapy, high dose brachytherapy, radical prostatectomy, рак предстательной железы, спасительная брахитерапия, высокодозная брахитерапия
File Description: application/pdf
Relation: https://oncourology.abvpress.ru/oncur/article/view/1717/1495; https://oncourology.abvpress.ru/oncur/article/downloadSuppFile/1717/1377; https://oncourology.abvpress.ru/oncur/article/downloadSuppFile/1717/1378; https://oncourology.abvpress.ru/oncur/article/downloadSuppFile/1717/1379; https://oncourology.abvpress.ru/oncur/article/downloadSuppFile/1717/1380; https://oncourology.abvpress.ru/oncur/article/downloadSuppFile/1717/1381; Siegel R.L., Miller K.D., Wagle N.S., Jemal A. Cancer statistics, 2023. CA Cancer J Clin 2023;73(1):17–48. DOI:10.3322/caac.21763; Huang J., On-Ting Chan E., Liu X. et al. Global trends of prostate cancer by age, and their associations with Gross Domestic Product (GDP), Human Development Index (HDI), smoking, and alcohol drinking. Clin Genitourin Cancer 2023;21(4):e261–70. DOI:10.1016/j.clgc.2023.02.003; Roberts M.J., Chatfield M.D., Hruby G. et al. Event-free survival after radical prostatectomy according to PSMA PET and EAU Biochemical Recurrence Risk Groups. BJU Int 2022;130(Suppl 3): 32–9. DOI:10.1111/bju.15762; Петровский Н.В., Глыбочко П.В., Аляев Ю.Г. и др. Локальный рецидив рака предстательной железы после радикальной простатэктомии. Урология 2017;(4):85–90. DOI:10.18565/urol.2017.4.85-90; Pound C.R., Partin A.W., Eisenberger M.A. et al. Natural history of progression after PSA elevation following radical prostatectomy. JAMA 1999;281(17):1591–7. DOI:10.1001/jama.281.17.1591; Moreira D.M., Presti J.C., Aronson W.J. et al. Natural history of persistently elevated prostate specific antigen after radical prostatectomy: results from the SEARCH database. J Urol 2009;182(5):2250–5. DOI:10.1016/j.juro.2009.07.022; Preisser F., Chun F.K.H., Pompe R.S. et al. Persistent prostatespecific antigen after radical prostatectomy and its impact on oncologic outcomes. Eur Urol 2019;76(1):106–14. DOI:10.1016/j.eururo.2019.01.048; Sugino F., Nakane K., Kawase M. et al. Biochemical recurrence after chemohormonal therapy followed by robot-assisted radical prostatectomy in very-high-risk prostate cancer patients. J Robot Surg 2023;17(5):2441–9. DOI:10.1007/s11701-023-01670-3; Le Guevelou J., Magne N., Counago F. et al. Stereotactic body radiation therapy after radical prostatectomy: current status and future directions. World J Urol 2023;41(11):3333–44. DOI:10.1007/s00345-023-04605-7; Falagario U.G., Abbadi A., Remmers S. et al. Biochemical recurrence and risk of mortality following radiotherapy or radical prostatectomy. JAMA Netw Open 2023;6(9):e2332900. DOI:10.1001/jamanetworkopen.2023.32900; Furrer M.A., Sathianathen N., Gahl B. et al. Oncological outcomes after attempted nerve-sparing radical prostatectomy (NSRP) in patients with high-risk prostate cancer are comparable to standard non-NSRP: a longitudinal long-term propensity-matched singlecentre study. BJU Int 2024;133(1):53–62. DOI:10.1111/bju.16126; Ploussard G., Fossati N., Wiegel T. et al. Management of persistently elevated prostate-specific antigen after radical prostatectomy: a systematic review of the literature. Eur Urol Oncol 2021;4(2): 150–69. DOI:10.1016/j.euo.2021.01.001; Buchser D., Gomez-Iturriaga A., Melcon I. et al. Salvage highdose-rate brachytherapy for histologically confirmed macroscopic local relapsed prostate cancer after radical prostatectomy. J Contemp Brachytherapy 2016;8(6):477–83. DOI:10.5114/jcb.2016.64441; Petersen P.M., Cook A.D., Sydes M.R. et al. Salvage radiation therapy after radical prostatectomy: analysis of toxicity by dosefractionation in the RADICALS-RT trial. Int J Radiat Oncol Biol Phys 2023;117(3):624–9. DOI:10.1016/j.ijrobp.2023.04.032; Benziane-Ouaritini N., Zilli T., Giraud A. et al. Prostatectomy bed image-guided dose-escalated salvage radiotherapy (SPIDER): an international multicenter retrospective study. Eur Urol Oncol 2023;6(4):390–8. DOI:10.1016/j.euo.2023.02.013; Thompson I.M., Valicenti R.K., Albertsen P. et al. Adjuvant and salvage radiotherapy after prostatectomy: AUA/ASTRO Guideline. J Urol 2013;190(2):441–9. DOI:10.1016/j.juro.2013.05.032; Morton G., McGuffin M., Chung H.T. et al. Prostate high doserate brachytherapy as monotherapy for low and intermediate risk prostate cancer: Efficacy results from a randomized phase II clinical trial of one fraction of 19 Gy or two fractions of 13.5 Gy. Radiother Oncol 2020;146:90–6. DOI:10.1016/j.radonc.2020.02.009; Johansson B., Olsén J.S., Karlsson L. et al. High-dose-rate brachytherapy as monotherapy for low- and intermediate-risk prostate cancer: long-term experience of Swedish single-center. J Contemp Brachytherapy 2021;13(3):245–53. DOI:10.5114/jcb.2021.105846; NCCN guidelines insights. Prostate cancer, version 1.2023. Available at: https://education.nccn.org.; EAU Guidelines. Edn. presented at the EAU Annual Congress Milan 2023. ISBN 978-94-92671-19-6.; Клинические рекомендации. Рак предстательной железы, 2021–2022–2023 (20.01.2023). Доступно по: http://disuria.ru.; Strouthos I., Karagiannis E., Zamboglou N., Ferentinos K. Highdose-rate brachytherapy for prostate cancer: Rationale, current applications, and clinical outcome. Cancer Rep (Hoboken) 2021;5(1):e1450. DOI:10.1002/cnr2.1450; Sargos P., Chabaud S., Latorzeff I. et al. Adjuvant radiotherapy versus early salvage radiotherapy plus short-term androgen deprivation therapy in men with localised prostate cancer after radical prostatectomy (GETUG-AFU 17): a randomised, phase 3 trial. Lancet Oncol 2020;21(10):1341–52. DOI:10.1016/S1470-2045(20)30454-X; Staal F.H.E., Janssen J., Brouwer C.L. et al. Phase III randomised controlled trial on PSMA PET/CT guided hypofractionated salvage prostate bed radiotherapy of biochemical failure after radical prostatectomy for prostate cancer (PERYTON-trial): study protocol. BMC Cancer 2022;22(1):416. DOI:10.1186/s12885-022-09493-5; Soror T., Melchert C., Rades D. et al. Salvage high-dose-rate interventional radiotherapy (brachytherapy) for locally relapsed prostate cancer after radical prostatectomy and subsequent external irradiation. J Contemp Brachytherapy 2023;15(3):159–65. DOI:10.5114/jcb.2023.128845; https://oncourology.abvpress.ru/oncur/article/view/1717
-
18Academic Journal
Authors: A. I. Alaverdyan, A. V. Govorov, A. О. Vasilyev, K. B. Kolontarev, E. A. Prilepskaya, D. Yu. Pushkar, А. И. Алавердян, А. В. Говоров, А. О. Васильев, К. Б. Колонтарев, Е. А. Прилепская, Д. Ю. Пушкарь
Source: Cancer Urology; Том 19, № 4 (2023); 125-130 ; Онкоурология; Том 19, № 4 (2023); 125-130 ; 1996-1812 ; 1726-9776
Subject Terms: робот-ассистированная хирургия, high-intensity focused ultrasound, HIFU, prostate cancer, robot-assisted surgery, высокоинтенсивный сфокусированный ультразвук, рак предстательной железы
File Description: application/pdf
Relation: https://oncourology.abvpress.ru/oncur/article/view/1678/1500; Состояние онкологической помощи населению России в 2021 году. Под ред. А.Д. Каприна, В.В. Старинского, А.О. Шахзадовой. М.: МНИОИ им. П.А. Герцена – филиал ФГБУ «НМИЦ радиологии» Минздрава России, 2022. 239 с.; Ahmed H.U., Akin O., Coleman J.A. et al. Transatlantic consensus group on active surveillance and focal therapy for prostate cancer. BJU Int 2012;109(11):1636–47. DOI:10.1111/j.1464-410X.2011.10633.x; Васильев А.В., Говоров А.В., Ширяев А.А. и др. Фокальная терапия рака предстательной железы: селекция пациентов, существующие ограничения и перспективы внедрения в клиническую практику. Онкоурология 2017;13(4):55–63. DOI:10.17650/1726-9776-2017-13-4-55-63; Левин В.П. Клинико-морфологическая и иммуногистохимическкая характеристика аденокарциномы предстательной железы в условиях HIFU-терапии. Дис. … канд. мед. наук. Новосибирск, 2019.; Chaussy C.G., Thuroff S.F. Robotic high-intensity focusedultrasound for prostate cancer: what have we learned in 15 years of clinical use? Curr Urol Rep 2011;12(3):180–7. DOI:10.1007/s11934-011-0184-2; EAU Guidelines. Edn. presented at the EAU Annual Congress Amsterdam 2022.; Пушкарь Д.Ю., Говоров А.В., Васильев А.О. и др. Реализация московской программы ранней диагностики и лечения рака предстательной железы. Труды научно-исследовательского института организации здравоохранения и медицинского менеджмента. Сборник научных трудов. Т. 5. М.: Государственное бюджетное учреждение города Москвы «Научно-исследовательский институт организации здравоохранения и медицинского менеджмента Департамента здравоохранения города Москвы», 2020. 128 с.; Mearini L., D’Urso L., Collura D. et al. High-intensity focused ultrasound for the treatment of prostate cancer: a prospective trial with long-term follow-up. Scand J Urol 2015;49(4):267–74. DOI:10.3109/21681805.2014.988174; https://oncourology.abvpress.ru/oncur/article/view/1678
-
19Academic Journal
Authors: M. I. Volkova, I. S. Al-Akel, Ya. V. Gridneva, R. I. Ryabinin, I. A. Pokataev, М. И. Волкова, И. С. Аль-Акел, Я. В. Гриднева, Р. И. Рябинин, И. А. Покатаев
Source: Cancer Urology; Том 20, № 3 (2024); 57-66 ; Онкоурология; Том 20, № 3 (2024); 57-66 ; 1996-1812 ; 1726-9776
Subject Terms: дистанционная лучевая терапия, non-metastatic prostate cancer, radical prostatectomy, external beam radiotherapy, неметастатический рак предстательной железы, радикальная простатэктомия
File Description: application/pdf
Relation: https://oncourology.abvpress.ru/oncur/article/view/1811/1557; Состояние онкологической помощи населению России в 2022 году. Под ред. А.Д. Каприна, В.В. Старинского, А.О. Шахзадовой. М.: МНИОИ им. П.А. Герцена – филиал ФГБУ «НМИЦ радиологии» Минздрава России, 2023. 239 с.; Ferlay J., Colombet M., Soerjomataram I. et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer 2019;144(8):1941–53. DOI:10.1002/ijc.31937; Носов Д.А., Волкова М.И., Гладков О.А. и др. Практические рекомендации по лекарственному лечению рака предстательной железы. Практические рекомендации RUSSCO, часть 1. Злокачественные опухоли 2023;13(3s2):640–60.; Houterman S., Janssen-Heijnen M.L., Verheij C.D. et al. Greater influence of age than co-morbidity on primary treatment and complications of prostate cancer patients: an in-depth population-based study. Prostate Cancer Prostatic Dis 2006;9(2):179–84. DOI:10.1038/sj.pcan.4500868; Bechis S.K., Carroll P.R., Cooperberg M.R. Impact of age at diagnosis on prostate cancer treatment and survival. J Clin Oncol 2011;29(2):235–41. DOI:10.1200/JCO.2010.30.2075; Preisser F., Mazzone E., Nazzani S. et al. Impact of age on perioperative outcomes at radical prostatectomy: a populationbased study. Eur Urol Focus 2020;6(6):1213–9. DOI:10.1016/j.euf.2018.12.006; Gurung P.M.S., Wang B., Hassig S. et al. Oncological and functional outcomes in patients over 70 years of age treated with robotic radical prostatectomy: a propensity-matched analysis. World J Urol 2021;39(4):1131–40. DOI:10.1007/s00345-020-03304-x; Yamada Y., Teshima T., Fujimura T. et al. Comparison of perioperative outcomes in elderly (age ≥75 years) vs. younger men undergoing robot-assisted radical prostatectomy. PLoS One 2020;15(6):e0234113. DOI:10.1371/journal.pone.0234113; Zelefsky M.J., Levin E.J., Hunt M. et al. Incidence of late rectal and urinary toxicities after three-dimensional conformal radiotherapy and intensity-modulated radiotherapy for localized prostate cancer. Int J Radiat Oncol Biol Phys 2008;70(4):1124–9. DOI:10.1016/j.ijrobp.2007.11.044; Xu N., Rossi P.J., Jani A.B. Toxicity analysis of dose escalation from 75.6 gy to 81.0 gy in prostate cancer. Am J Clin Oncol 2011;34(1):11–5. DOI:10.1097/COC.0b013e3181cae8c6; Eade T.N., Hanlon A.L., Horwitz E.M. et al. What dose of externalbeam radiation is high enough for prostate cancer? Int J Radiat Oncol Biol Phys 2007;68(3):682–9. DOI:10.1016/j.ijrobp.2007.01.008; Kumar S., Shelley M., Harrison C. et al. Neo-adjuvant and adjuvant hormone therapy for localised and locally advanced prostate cancer. Cochrane Database Syst Rev 2006;2006(4):CD006019. DOI:10.1002/14651858.CD006019.pub2; Spratt D.E., Malone S., Roy S. et al. Prostate radiotherapy with adjuvant androgen deprivation therapy (ADT) improves metastasis-free survival compared to neoadjuvant ADT: an individual patient meta-analysis. J Clin Oncol 2021;39(2):136–44. DOI:10.1200/JCO.20.02438; NCCN guideline for prostate cancer. Available at: https://www.nccn.org/home; Shipley W.U., Seiferheld W., Lukka H.R. et al. Radiation with or without antiandrogen therapy in recurrent prostate cancer. N Engl J Med 2017;376(5):417–28. DOI:10.1056/NEJMoa1607529; D’Amico A.V., Chen M.H., Renshaw A.A. et al. Androgen suppression and radiation vs radiation alone for prostate cancer: a randomized trial. JAMA 2008;299(3):289–95. DOI:10.1001/jama.299.3.289; Denham J.W., Steigler A., Lamb D.S. et al. Short-term neoadjuvant androgen deprivation and radiotherapy for locally advanced prostate cancer: 10-year data from the TROG 96.01 randomised trial. Lancet Oncol 2011;12(5):451–9. DOI:10.1016/S1470-2045(11)70063-8; Jones C.U., Hunt D., McGowan D.G. et al. Radiotherapy and short-term androgen deprivation for localized prostate cancer. N Engl J Med 2011;365(2):107–18. DOI:10.1056/NEJMoa1012348; Horwitz E.M., Bae K., Hanks G.E. et al. Ten-year follow-up of radiation therapy oncology group protocol 92-02: a phase III trial of the duration of elective androgen deprivation in locally advanced prostate cancer. J Clin Oncol 2008;26(15):2497–504. DOI:10.1200/JCO.2007.14.9021; Bolla M., de Reijke T.M., Van Tienhoven G. et al. Duration of androgen suppression in the treatment of prostate cancer. N Engl J Med 2009;360(24):2516–27. DOI:10.1056/NEJMoa0810095; Zapatero A., Guerrero A., Maldonado X. et al. High-dose radiotherapy with short-term or long-term androgen deprivation in localised prostate cancer (DART01/05 GICOR): a randomised, controlled, phase 3 trial. Lancet Oncol 2015;16(6):320–7. DOI:10.1016/S1470-2045(15)70272-X; Kishan A.U., Wang X., Seiferheld W. et al. Association of Gleason grade with androgen deprivation therapy duration and survival outcomes: a systematic review and patient-level meta-analysis. JAMA Oncol 2018;5(1):91–6. DOI:10.1001/jamaoncol.2018.3732; Nabid A., Carrier N., Martin A.G. et al. Duration of androgen deprivation therapy in high-risk prostate cancer: a randomized phase III trial. Eur Urol 2018;74(4):432–41. DOI:10.1016/j.eururo.2018.06.018; Denham J.W., Joseph D., Lamb D.S. et al. Short-term androgen suppression and radiotherapy versus intermediate-term androgen suppression and radiotherapy, with or without zoledronic acid, in men with locally advanced prostate cancer (TROG 03.04 RADAR): 10-year results from a randomised, phase 3, factorial trial. Lancet Oncol 2019;20(2):267–81. DOI:10.1016/S1470-2045(18)30757-5; Hamdy F.C., Donovan J.L., Lane J.A. et al.; ProtecT Study Group. Fifteen-year outcomes after monitoring, surgery, or radiotherapy for prostate cancer. N Engl J Med 2023;388(17):1547–58. DOI:10.1056/NEJMoa2214122; Duchesne G.M., Woo H.H., Bassett J.K. et al. Timing of androgendeprivation therapy in patients with prostate cancer with a rising PSA (TROG 03.06 and VCOG PR 01-03 [TOAD]): a randomised, multicentre, non-blinded, phase 3 trial. Lancet Oncol 2016;17(6):727–37.; Boorjian S.A., Thompson R.H., Tollefson M.K. et al. Long-term risk of clinical progression after biochemical recurrence following radical prostatectomy: the impact of time from surgery to recurrence. Eur Urol 2011;59(6):893–9. DOI:10.1016/j.eururo.2011.02.026; https://oncourology.abvpress.ru/oncur/article/view/1811
-
20Academic Journal
Authors: E. A. Nikolaeva, A. S. Krylov, A. D. Ryzhkov, T. M. Geliashvili, A. V. Pavlova, A. Yu. Goryainova, R. A. Murashko, Е. А. Николаева, А. С. Крылов, А. Д. Рыжков, Т. М. Гелиашвили, А. В. Павлова, А. Ю. Горяинова, Р. А. Мурашко
Source: Cancer Urology; Том 20, № 2 (2024); 74-86 ; Онкоурология; Том 20, № 2 (2024); 74-86 ; 1996-1812 ; 1726-9776
Subject Terms: радионуклидная терапия, metastatic castration-resistant prostate cancer, quantitative single-photon emission computed tomography/computed tomography, radium-223 dichloride, radionuclide therapy, метастатический кастрационно-резистентный рак предстательной железы, количественная однофотонная эмиссионная компьютерная томография, совмещенная с компьютерной томографией, дихлорид радия-223
File Description: application/pdf
Relation: https://oncourology.abvpress.ru/oncur/article/view/1762/1547; Tannock I.F., de Wit R., Berry W.R. et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med 2004;351(15):1502–12. DOI:10.1056/NEJMoa040720; Pezaro C., Omlin A., Lorente D. et al. Visceral disease in castration-resistant prostate cancer. Eur Urol 2014;65(2):270–3. DOI:10.1016/j.eururo.2013.10.055; Smith H.S. Painful osseous metastases. Pain Physician 2011;14(4):E373–403.; Coleman R.E. Clinical features of metastatic bone disease and risk of skeletal morbidity. Clin Cancer Res 2006;12(20 Pt 2):6243s–9s. DOI:10.1158/1078-0432.CCR-06-0931; Tait C., Moore D., Hodgson C. et al. Quantification of skeletal metastases in castrate-resistant prostate cancer predicts progressionfree and overall survival. BJU Int 2014;114(6b):E70–3. DOI:10.1111/bju.12717; Riihimäki M., Thomsen H., Brandt A. et al. What do prostate cancer patients die of? Oncologist 2011;16(2):175–81. DOI:10.1634/theoncologist.2010-0338; Cornford P., Bellmunt J., Bolla M. et al. EAU-ESTRO-SIOG guidelines on prostate cancer. Part II: treatment of relapsing, metastatic, and castration-resistant prostate cancer. Eur Urol 2017;71(4):630–42. DOI:10.1016/j.eururo.2016.08.002; Parker C., Nilsson S., Heinrich D. et al. Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med 2013;369(3):213–23. DOI:10.1056/NEJMoa1213755; Sartor O., Coleman R., Nilsson S. et al. Effect of radium-223 dichloride on symptomatic skeletal events in patients with castration-resistant prostate cancer and bone metastases: results from a phase 3, double-blind, randomised trial. Lancet Oncol 2014;15(7):738–46. DOI:10.1016/S1470-2045(14)70183-4; Hoskin P., Sartor O., O’Sullivan J.M. et al. Efficacy and safety of radium-223 dichloride in patients with castration-resistant prostate cancer and symptomatic bone metastases, with or without previous docetaxel use: a prespecified subgroup analysis from the randomised, double-blind, phase 3 ALSYMPCA trial. Lancet Oncol 2014; 15(12):1397–406. DOI:10.1016/S1470-2045(14)70474-7; Nilsson S., Cislo P., Sartor O. et al. Patient-reported quality-of-life analysis of radium-223 dichloride from the phase III ALSYMPCA study. Ann Oncol 2016;27(5):868–74. DOI:10.1093/annonc/mdw065; Parker C., Finkelstein S.E., Michalski J.M. et al. Efficacy and safety of radium-223 dichloride in symptomatic castration-resistant prostate cancer patients with or without baseline opioid use from the phase 3 ALSYMPCA trial. Eur Urol 2016;70(5):875–83. DOI:10.1016/j.eururo.2016.06.002; Николаева Е.А., Тарачкова Е.В., Шейх Ж.В. и др. Обзор методов визуализации для оценки ответа на лечение метастазов в костях при раке предстательной железы и молочной железы. Кремлевская медицина. Клинический вестник 2022;3:107–14. DOI:10.26269/tvby-7e26; Helyar V., Mohan H.K., Barwick T. et al. The added value of multislice SPECT/CT in patients with equivocal bony metastasis from carcinoma of the prostate. Eur J Nucl Med Mol Imaging 2010;37(4):706–13. DOI:10.1007/s00259-009-1334-3; Vogelzang N.J., Coleman R.E., Michalski J.M. et al. Hematologic safety of radium-223 dichloride: baseline prognostic factors associated with myelosuppression in the ALSYMPCA trial. Clin Genitourin Cancer 2017;15(1):42–52.e8. DOI:10.1016/j.clgc.2016.07.027; Etchebehere E.C., Araujo J.C., Fox P.S. et al. Prognostic factors in patients treated with 223Ra: the role of skeletal tumor burden on baseline 18F-fluoride PET/CT in predicting overall survival. J Nucl Med 2015;56(8):1177–84. DOI:10.2967/jnumed.115.158626; Ulmert D., Kaboteh R., Fox J.J. et al. A novel automated platform for quantifying the extent of skeletal tumour involvement in prostate cancer patients using the Bone Scan Index. Eur Urol 2012;62(1): 78–84. DOI:10.1016/j.eururo.2012.01.037; Fosbøl M.Ø., Petersen P.M., Kjaer A., Mortensen J. 223Ra therapy of advanced metastatic castration-resistant prostate cancer: quantitative assessment of skeletal tumor burden for prognostication of clinical outcome and hematologic toxicity. J Nucl Med 2018;59(4):596–602. DOI:10.2967/jnumed.117.195677; Shariftabrizi A., Kothari S., George S. et al. Optimization of radium-223 treatment of castration-resistant prostate cancer based on the burden of skeletal metastasis and clinical parameters. Oncologist 2023;28(3):246–51. DOI:10.1093/oncolo/oyac245; Николаева Е.А., Крылов А.С., Рыжков А.Д. и др. Количественная оценка методом ОФЭКТ/КТ эффективности радионуклидной терапии хлоридом радия-223 костных метастазов при метастатическом кастрационно-резистентном раке предстательной железы. Онкологический журнал: лучевая диагностика, лучевая терапия 2022;5(3):29–42. DOI:10.37174/2587-7593-2022-5-3-29-42; Shore N.D. Radium-223 dichloride for metastatic castrationresistant prostate cancer: the urologist’s perspective. Urology 2015;85(4):717–24. DOI:10.1016/j.urology.2014.11.031; Gillessen S., Omlin A., Attard G. et al. Management of patients with advanced prostate cancer: recommendations of the St Gallen Advanced Prostate Cancer Consensus Conference (APCCC) 2015. Ann Oncol 2015;26(8):1589–604. DOI:10.1093/annonc/mdv257; CTCAE. National Cancer Institute Enterprise Vocabulary Services website. Available at: https://evs.nci.nih.gov/ftp1/CTCAE/About.html (accessed 15 December 2023).; Asselah J., Sperlich C. Post-docetaxel options for further survival benefit in metastatic castration-resistant prostate cancer: questions of choice. Can Urol Assoc J 2013;7(1–2 Suppl 1):S11–7. DOI:10.5489/cuaj.274; Sathianathen N.J., Lamb A., Nair R. et al. Updates of prostate cancer staging: prostate-specific membrane antigen. Investig Clin Urol 2016;57(Suppl 2):S147–54. DOI:10.4111/icu.2016.57.S2.S147; Lowrance W.T., Roth B.J., Kirkby E. et al. Castration-resistant prostate cancer: AUA guideline amendment 2015. J Urol 2016;195(5):1444–52. DOI:10.1016/j.juro.2015.10.086; Parker C., Gillessen S., Heidenreich A., Horwich A. ESMO Guidelines Committee. Cancer of the prostate: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 2015;26(Suppl 5):v69–77. DOI:10.1093/annonc/mdv222; Saad F., Chi K.N., Finelli A. et al. The 2015 CUA-CUOG Guidelines for the management of castration-resistant prostate cancer (CRPC). Can Urol Assoc J 2015;9(3–4):90–6. DOI:10.5489/cuaj.2526; Носов Д.А., Волкова М.И., Гладков О.А. и др. Практические рекомендации по лечению рака предстательной железы. Злокачественные опухоли: Практические рекомендации RUSSCO 2021;11(3s2):540–55. DOI:10.18027/2224-5057-2021-11-3s2-33; Smith M., Parker C., Saad F. et al. Addition of radium-223 to abiraterone acetate and prednisone or prednisolone in patients with castration-resistant prostate cancer and bone metastases (ERA 223): a randomised, double-blind, placebo-controlled, phase 3 trial [published correction appears in Lancet Oncol 2019;20(10):e559]. Lancet Oncol 2019;20(3):408–19. DOI:10.1016/S1470-2045(18)30860-X; Choudhury A.D., Kwak L., Cheung A. et al. Randomized phase II study evaluating the addition of pembrolizumab to radium-223 in metastatic castration-resistant prostate cancer. Journal of Clinical Oncology 2021;39(6_suppl):98. DOI:10.1200/JCO.2021.39.6_suppl.98; Sternberg C.N., Saad F., Graff J.N. et al. A randomised phase II trial of three dosing regimens of radium-223 in patients with bone metastatic castration-resistant prostate cancer. Ann Oncol 2020;31(2):257–65. DOI:10.1016/j.annonc.2019.10.025; Carles J., Alonso-Gordoa T., Mellado B. et al. Radium-223 for patients with metastatic castration-resistant prostate cancer with asymptomatic bone metastases progressing on first-line abiraterone acetate or enzalutamide: a single-arm phase II trial. Eur J Cancer 2022;173:317–26. DOI:10.1016/j.ejca.2022.06.057; Marshall C.H., Fu W., Wang H. et al. Randomized phase II trial of sipuleucel-T with or without radium-223 in men with bonemetastatic castration-resistant prostate cancer. Clin Cancer Res 2021;27(6):1623–30. DOI:10.1158/1078-0432.CCR-20-4476; Morris M.J., Loriot Y., Sweeney C.J. et al. Radium-223 in combination with docetaxel in patients with castration-resistant prostate cancer and bone metastases: a phase 1 dose escalation/ randomised phase 2a trial. Eur J Cancer 2019;114:107–16. DOI:10.1016/j.ejca.2019.04.007; Darolutamide with radium-223 or placebo and the effect on radiological Progression-free survival for patients with mCSPC (CARE). ClinicalTrials.gov. ID NCT05771896. Available at: https://clinicaltrials.gov/study/NCT05771896; Kostos L., Buteau J.P., Yeung T. et al. AlphaBet: combination of radium-223 and [177Lu]Lu-PSMA-I&T in men with metastatic castration-resistant prostate cancer (clinical trial protocol). Front Med (Lausanne) 2022;9:1059122. DOI:10.3389/fmed.2022.1059122; Pezaro C.J., Omlin A., Lorente D. et al. Visceral disease in castration-resistant prostate cancer. Eur Urol 2014;65:270–3. DOI:10.1016/j.eururo.2013.10.055; Bosch D., van der Velden K.J.M., Oving I.M. et al. The impact of baseline PSMA PET/CT vs. CT on outcomes of Radium-223 therapy in mCRPC patients. Annals of Oncology 2023;34(Suppl 2): 987–8. DOI:10.1016/j.annonc.2023.09.2771; Carles J., Castellano D., Méndez-Vidal M.J. et al. Circulating tumor Cells as a biomarker of survival and response to Radium-223 therapy: experience in a cohort of patients with metastatic castration-resistant prostate cancer. Clin Genitourin Cancer 2018;16(6):e1133–9. DOI:10.1016/j.clgc.2018.07.013; Iizuka J. Evaluating radium-223 response in metastatic castrationresistant prostate cancer with imaging. Asia Pac J Clin Oncol 2018;14(Suppl 5):16–23. DOI:10.1111/ajco.13058; Scher H.I., Morris M.J., Stadler W.M. et al. Prostate cancer clinical trials working group 3. Trial design and objectives for castrationresistant prostate cancer: updated recommendations from the prostate cancer clinical trials working group 3. J Clin Oncol 2016;34(12):1402–18. DOI:10.1200/JCO.2015.64.2702; https://oncourology.abvpress.ru/oncur/article/view/1762