-
1Academic Journal
Authors: A. A. Zagidullina, L. A. Dzhambulatova, M. A. Shatueva, T. B. Dongak, Y. S. Laubakh, D. S. Shakirova, A. S. Golantsev, Kh. R. Payzulaeva, D. P. Yastrebova, A. M. Aksenov, E. S. Goncharova, M. A. Ozhereleva, D. G. Sargsyan, А. А. Загидуллина, Л. А. Джамбулатова, М. А. Шатуева, Т. Б. Донгак, Я. С. Лаубах, Д. С. Шакирова, А. С. Голанцев, Х. Р. Пайзулаева, Д. П. Ястребова, А. М. Аксенов, Е. С. Гончарова, М. А. Ожерельева, Д. Г. Саргсян
Contributors: The authors declare no funding, Авторы заявляют об отсутствии финансовой поддержки
Source: Obstetrics, Gynecology and Reproduction; Vol 19, No 3 (2025); 389-407 ; Акушерство, Гинекология и Репродукция; Vol 19, No 3 (2025); 389-407 ; 2500-3194 ; 2313-7347
Subject Terms: гонадотоксичность, BC, fertility preservation, oocyte cryopreservation, embryo cryopreservation, ovarian tissue cryopreservation, gonadotoxicity, РМЖ, сохранение фертильности, криоконсервация яйцеклеток, криоконсервация эмбрионов, криоконсервация ткани яичников
File Description: application/pdf
Relation: https://www.gynecology.su/jour/article/view/2387/1347; Мерабишвили В.М., Семиглазов В.Ф., Комяхов А.В. и др. Состояние онкологической помощи в России: рак молочной железы. Эпидемиология и выживаемость больных. Влияние эпидемии бета-варианта коронавируса SARS-CoV-2 (клинико-популяционное исследование). Опухоли женской репродуктивной системы. 2023;19(3):16–24. https://doi.org/10.17650/1994-4098-2023-19-3-16-24.; Тюляндин С.А., Стенина М.Б., Фролова М.А. Практические инструменты, облегчающие выбор адъювантной лекарственной терапии у больных операбельным люминальным HER2-негативным раком молочной железы. Злокачественные опухоли. 2024;14(2):51–7. https://doi.org/10.18027/2224-5057-2024-003.; Павлова В.И., Белая Ю.А., Воронцов А.Ю. и др. 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. https://doi.org/10.17650/1994-4098-2023-19-2-54-60.; Ахапкина Е.С., Макиева М.И., Хохлова С.В. и др. Особенности состояния здоровья детей, рожденных у матерей с раком молочной железы во время беременности: обзор литературы. Неонатология: новости, мнения, обучение. 2023;11(4):21–5. https://doi.org/10.33029/2308-2402-2023-11-4-21-25.; Llarena N.C., Estevez S.L., Tucker S.L., Jeruss J.S. Impact of fertility concerns on tamoxifen initiation and persistence. J Natl Cancer Inst. 2015;107(10):djv202. https://doi.org/10.1093/jnci/djv202.; Oktay K., Harvey B.E., Partridge A.H. et al. Fertility preservation in patients with cancer: ASCO Clinical Practice Guideline Update. J Clin Oncol. 2018;36(19):1994–2001. https://doi.org/10.1200/JCO.2018.78.1914.; Vuković P., Kasum M., Raguž J. et al. Fertility preservation in young women with early-stage breast cancer. Acta Clin Croat. 2019;58(1):147–56. https://doi.org/10.20471/acc.2019.58.01.19.; Partridge A.H., Pagani O., Abulkhair O. et al. First international consensus guidelines for breast cancer in young women (BCY1). Breast. 2014;23(3):209–20. https://doi.org/10.1016/j.breast.2014.03.011.; Lambertini M., Del Mastro L., Pescio M.C. et al. Cancer and fertility preservation: international recommendations from an expert meeting. BMC Med. 2016;14:1. https://doi.org/10.1186/s12916-015-0545-7.; Переводчикова Н.И., Стенина М.Б. Современные возможности индивидуализации лекарственной терапии рака молочной железы. Российский онкологический журнал. 2016;21(1–2):18–25. https://doi.org/10.18821/1028-9984-2015-21-1-18-25.; Вартанян Э.В., Доброхотова Ю.Э., Девятова Е.А., Цатурова К.А. Сохранение женской фертильности при онкологических заболеваниях. Проблемы репродукции. 2020;26(4):68–76. https://doi.org/10.17116/repro20202604168.; Castillo C., Camejo N. Impact of breast cancer treatments on fertility and the importance of timing for a fertility preservation intervention. Rev Senol Patol Mam. 2022;35(4):305–11. https://doi.org/10.1016/j.senol.2021.08.004.; Mauri D., Gazouli I., Zarkavelis G. et al. Chemotherapy associated ovarian failure. Front Endocrinol. 2020;11:572388. https://doi.org/10.3389/fendo.2020.572388.; Wang S., Pei L., Hu T. et al. Protective effect of goserelin on ovarian reserve during (neo)adjuvant chemotherapy in young breast cancer patients: a prospective cohort study in China. Hum Reprod. 2021;36(4):976–86. https://doi.org/10.1093/humrep/deaa349.; Blumenfeld Z. Fertility preservation in women with malignancy: future endeavors. Clin Med Insights Reprod Health. 2019;13:1179558119872490. https://doi.org/10.1177/1179558119872490.; Çelebi F., Ordu Ç., Ilgün S. et al. The effect of systemic chemotherapy on ovarian function: a prospective clinical trial. Eur J Breast Health. 2020;16(3):177–82. https://doi.org/10.5152/ejbh.2020.5114.; Зикиряходжаев А.Д., Новикова О.В., Рассказова Е.А. Возможность беременностей и родов после лечения рака молочной железы. Архив акушерства и гинекологии имени В.Ф. Снегирева. 2017;4(3):149–53. https://doi.org/10.18821/2313-8726-2017-4-3-149-153.; Белоусов Д.Ю., Чеберда А.Е. Адъювантная гормональная терапия на ранней стадии эстрогенположительного рака молочной железы: систематический обзор и сетевой метаанализ. Пациентоориентированная медицина и фармация. 2023;1(2):75–84. https://doi.org/10.37489/2949-1924-0017.; Имянитов Е.Н. Эволюция системного лечения гормонозависимого рака молочной железы: от чередования препаратов к комбинированной терапии. Опухоли женской репродуктивной системы. 2016;12(2):46–51. https://doi.org/10.17650/1994-4098-2016-12-2-46-51.; Tremont A., Lu J., Cole J.T. Endocrine therapy for early breast cancer: updated review. Ochsner J. 2017;17(4):405–11.; Назаренко Т.А., Пароконная А.А., Шарипова Н.Ю. Применение ингибиторов ароматазы у больных раком молочной железы в программах вспомогательных репродуктивных технологий. Проблемы репродукции. 2012;(2):72–6.; Савельева М.И., Голубенко Е.О., Созаева Ж.А. и др. Анализ осложнений эндокринотерапии тамоксифеном при раке молочной железы: клинические и фармакогенетические аспекты. Современная oнкология. 2022;24(3):361–7. https://doi.org/10.26442/18151434.2022.3.201783.; Shandley L.M., Spencer J.B., Fothergill A. et al. Impact of tamoxifen therapy on fertility in breast cancer survivors. Fertil Steril. 2017;107(1):243–252.e5. https://doi.org/10.1016/j.fertnstert.2016.10.020.; Kim H.J., Noh W.C., Nam S.J. et al. Five-year changes in ovarian function restoration in premenopausal patients with breast cancer taking tamoxifen after chemotherapy: an ASTRRA study report. Eur J Cancer. 2021;151:190–200. https://doi.org/10.1016/j.ejca.2021.03.017.; Шевченко Ю.А., Кузнецова М.С., Христин А.А. и др. Современная терапия рака молочной железы: от тамоксифена до Т-клеточной инженерии. Сибирский онкологический журнал. 2022;21(5):109–22. https://doi.org/10.21294/1814-4861-2022-21-5-109-122.; Mehta P., Bothra S.J. PARP inhibitors in hereditary breast and ovarian cancer and other cancers: а review. Adv Genet. 2021;108:35–80. https://doi.org/10.1016/bs.adgen.2021.08.002.; Li J., Li Q., Zhang L. et al. Poly-ADP-ribose polymerase (PARP) inhibitors and ovarian function. Biomed Pharmacother. 2023;157:114028. https://doi.org/10.1016/j.biopha.2022.114028.; Артамонова Е.В., Коваленко Е.И., Болотина Л.В. и др. Ингибиторы CDK4/6 в терапии метастатического рака молочной железы. Выбор оптимальной терапии эстроген-рецептор-положительного HER2-отрицательного метастатического рака молочной железы: общероссийский анализ предпочтений врачей («Прометей») – первые результаты опроса. Опухоли женской репродуктивной системы. 2020;16(3):37–45. https://doi.org/10.17650/1994-4098-2020-16-3-37-45.; Dong F., Meng T.G., Li J. et al. Inhibition of CDK4/6 kinases causes production of aneuploid oocytes by inactivating the spindle assembly checkpoint and accelerating first meiotic progression. Biochim Biophys Acta Mol Cell Res. 2021;1868(7):119044. https://doi.org/10.1016/j.bbamcr.2021.119044.; Scavone G., Ottonello S., Blondeaux E. et al. The role of cyclin-dependent kinases (CDK) 4/6 in the ovarian tissue and the possible effects of their exogenous inhibition. Cancers. 2023;15(20):4923. https://doi.org/10.3390/cancers15204923.; Catlin N.R., Bowman C.J., Engel S.M et al. Reproductive and developmental toxicity assessment of palbociclib, a CDK4/6 inhibitor, in Sprague-Dawley rats and New Zealand White rabbits. Reprod Toxicol. 2019;88:76–84. https://doi.org/10.1016/j.reprotox.2019.07.016.; Болотина Л.В. Стратегия лекарственной терапии метастатического рака молочной железы в условиях расширения палитры противоопухолевых средств. Вопросы онкологии. 2024;70(2):248–58. https://doi.org/10.37469/0507-3758-2024-70-2-248-258.; Garutti M., Lambertini M., Puglisi F. Checkpoint inhibitors, fertility, pregnancy, and sexual life: a systematic review. ESMO Open. 2021;6(5):100276. https://doi.org/10.1016/j.esmoop.2021.100276.; Пигарова Е.А., Шутова А.С., Дзеранова Л.К. Коррекция эндокринных осложнений онкоиммунотерапии. Ожирение и метаболизм. 2022;19(4):418–30. https://doi.org/10.14341/omet12828.; Нуралиева Н.Ф., Трошина Е.А., Мельниченко Г.А. Поражение желез внутренней секреции как осложнение иммунотерапии в практике онколога. Клиническая и экспериментальная тиреоидология. 2018;14(4):174–82. https://doi.org/10.14341/ket9875.; Charmsaz S., Scott A.M., Boyd A.W. Targeted therapies in hematological malignancies using therapeutic monoclonal antibodies against Eph family receptors. Exp Hematol. 2017;54:31–9. https://doi.org/10.1016/j.exphem.2017.07.003.; Mandó P., Waisberg F., Pasquinelli R. et al. HER2-directed therapy in advanced breast cancer: benefits and risks. Onco Targets Ther. 2023;16:115–32. https://doi.org/10.2147/OTT.S335934.; Артамонова Е.В., Лубенникова Е.В. Оптимальный выбор неоадъювантной терапии HER2-положительного рака молочной железы. Анализ предпочтений врачей в Российской Федерации. Медицинский алфавит. 2023;(27):7–12. https://doi.org/10.33667/2078-5631-2023-27-7-12.; Levi M., Goshen-Lago T., Yerushalmi R. et al. Anti-HER2/neu antibody reduces chemotherapy-induced ovarian toxicity-from bench to bedside. Biomedicines. 2020;8(12):577. https://doi.org/10.3390/biomedicines8120577.; Lambertini M., Campbell C., Bines J. et al. Adjuvant anti-HER2 therapy, treatment-related amenorrhea, and survival in premenopausal HER2-positive early breast cancer patients. J Natl Cancer Inst. 2019;111(1):86–94. https://doi.org/10.1093/jnci/djy094.; Адамян Л.В., Носов В.Б., Степанян А.А. Лечение, сохраняющее фертильность у онкологических пациентов: чем мы можем помочь в XXI веке? Проблемы репродукции. 2024;30(1):26–63. https://doi.org/10.17116/repro20243001126.; Poorvu P.D., Hu J., Zheng Y. et al. Treatment-related amenorrhea in a modern, prospective cohort study of young women with breast cancer. NPJ Breast Cancer. 2021;7(1):99. https://doi.org/10.1038/s41523-021-00307-8.; Дашян Г.А., Семиглазов В.Ф., Криворотько П.В. и др. Новые перспективы применения препарата Кадсила® при раке молочной железы. Опухоли женской репродуктивной системы. 2015;11(4):46–52. https://doi.org/10.17650/1994-4098-2015-11-4-46-52.; Cui W., Francis P.A., Loi S. et al. Assessment of ovarian function in phase III (neo)adjuvant breast cancer clinical trials: a systematic evaluation. J Natl Cancer Inst. 2021;113(12):1770–8. https://doi.org/10.1093/jnci/djab111.; Афонин Г.В., Рагулин Ю.А., Гулидов И.А. и др. Эффективность адъювантной лучевой терапии в режиме гипофракционирования у больных операбельным раком молочной железы. Сибирский онкологический журнал. 2018;17(5):37–44. https://doi.org/10.21294/1814-4861-2018-17-5-37-44.; Beyer S., Sandu A., White J. Impact and timing of breast cancer radiation therapy and fertility preservation. Curr Breast Cancer Rep. 2020;12(4):375–80. https://doi.org/10.1007/s12609-020-00394-9.; Краснопольская К.В., Крстич Е.В., Горская О.С. и др. Место агонистов гонадотропин-рилизинг-гормонов в репродуктивной медицине. Проблемы репродукции. 2012;(2):65–7.; Пустотина О.А. Агонисты гонадотропин-рилизинг-гормона и add-back-терапия. Российский вестник акушера-гинеколога. 2023;23(2):63–9. https://doi.org/10.17116/rosakush20232302163.; Li Z.Y., Dong Y.L., Cao X.Z. et al. Gonadotropin-releasing hormone agonists for ovarian protection during breast cancer chemotherapy: a systematic review and meta-analysis. Menopause. 2022;29(9):1093–100. https://doi.org/10.1097/GME.0000000000002019.; Lambertini M., Boni .L, Michelotti A. et al. Ovarian suppression with triptorelin during adjuvant breast cancer chemotherapy and long-term ovarian function, pregnancies, and disease-free survival: a randomized clinical trial. JAMA. 2015;314(24):2632–40. https://doi.org/10.1001/jama.2015.17291.; Краснопольская К.В., Назаренко Т.А., Здановский В.М. и др. Влияние количественных параметров фолликулогенеза при контролируемой стимуляции яичников на эффективность ЭКО. Проблемы репродукции. 2017;23(3):55–61. https://doi.org/10.17116/repro201723355-61.; Park S.Y., Jeong K., Cho E.H., Chung H.W. Controlled ovarian hyperstimulation for fertility preservation in women with breast cancer: practical issues. Clin Exp Reprod Med. 2021;48(1):1–10. https://doi.org/10.5653/cerm.2020.03594.; Cavagna F., Pontes A., Cavagna M. et al. Specific protocols of controlled ovarian stimulation for oocyte cryopreservation in breast cancer patients. Curr Oncol. 2018;25(6):e527–e532. https://doi.org/10.3747/co.25.3889.; Baig A.S., Camuñas N.G., Sánchez P.P. et al. Controlled ovarian stimulation initiated at different phases of the menstrual cycle for fertility preservation in oncological patients: a retrospective study. Reprod Sci. 2023;30(8):2547–53. https://doi.org/10.1007/s43032-023-01175-2.; Cascante S.D., Berkeley A.S., Licciardi F. et al. Planned oocyte cryopreservation: the state of the ART. Reprod Biomed Online. 2023;47(6):103367. https://doi.org/10.1016/j.rbmo.2023.103367.; Lambertini M., Peccatori F.A., Demeestere I. et al. Fertility preservation and post-treatment pregnancies in post-pubertal cancer patients: ESMO Clinical Practice Guidelines. Ann Oncol. 2020;31(12):1664–78. https://doi.org/10.1016/j.annonc.2020.09.006.; Mahajan N. Fertility preservation in female cancer patients: an overview. J Hum Reprod Sci. 2015;8(1):3–13. https://doi.org/10.4103/0974-1208.153119.; Rosenberg E., Fredriksson A., Einbeigi Z. et al. No increased risk of relapse of breast cancer for women who give birth after assisted conception. Hum Reprod Open. 2019;2019(4):hoz039. https://doi.org/10.1093/hropen/hoz039.; Arecco L., Blondeaux E., Bruzzone M. et al. Safety of fertility preservation techniques before and after anticancer treatments in young women with breast cancer: a systematic review and meta-analysis. Hum Reprod. 2022;37(5):954–68. https://doi.org/10.1093/humrep/deac035.; Fraison E., Huberlant S., Labrune E. et al. Live birth rate after female fertility preservation for cancer or haematopoietic stem cell transplantation: a systematic review and meta-analysis of the three main techniques; embryo, oocyte and ovarian tissue cryopreservation. Hum Reprod. 2023;38(3):489–502. https://doi.org/10.1093/humrep/deac249.; Da Luz C.M., Caetano M.A., Berteli T.S. et al. The impact of oocyte vitrification on offspring: a systematic review. Reprod Sci. 2022;29(11):3222–34. https://doi.org/10.1007/s43032-022-00868-4.; Юпатов Е.Ю., Курманбаев Т.Е., Шмидт А.А. и др. Криоконсервация репродуктивной ткани – возможность сохранения фертильности (обзор литературы). Проблемы репродукции. 2020;26(5):99–106. https://doi.org/10.17116/repro20202605199.; Рухляда Н.Н., Казанцев В.А. Вопросы онкофертильности – обзор современных методов криоконсервации яичниковой ткани. Проблемы репродукции. 2018;24(1):53–6. https://doi.org/10.17116/repro201824153-56.; Albamonte M.I., Vitullo A.D. Preservation of fertility in female and male prepubertal patients diagnosed with cancer. J Assist Reprod Genet. 2023;40(12):2755–67. https://doi.org/10.1007/s10815-023-02945-2.; Dolmans M.M., von Wolff M., Poirot C. et al. Transplantation of cryopreserved ovarian tissue in a series of 285 women: a review of five leading European centers. Fertil Steril. 2021;115(5):1102–15. https://doi.org/10.1016/j.fertnstert.2021.03.008.; Boutas I., Kontogeorgi A., Koufopoulos N. et al. Breast cancer and fertility preservation in young female patients: a systematic review of the literature. Clin Pract. 2023;13(6):1413–26. https://doi.org/10.3390/clinpract13060127.; Sánchez-Serrano M., Crespo J., Mirabet V. et al. Twins born after transplantation of ovarian cortical tissue and oocyte vitrification. Fertil Steril. 2010;93(1):268.e11–3. https://doi.org/10.1016/j.fertnstert.2009.09.046.; Jensen A.K., Macklon K.T., Fedder J. et al. 86 successful births and 9 ongoing pregnancies worldwide in women transplanted with frozen-thawed ovarian tissue: focus on birth and perinatal outcome in 40 of these children. J Assist Reprod Genet. 2017;34(3):325–36. https://doi.org/10.1007/s10815-016-0843-9.; Klock S.C., Lindheim S.R. Disposition of unused cryopreserved embryos: opportunities and liabilities. Fertil Steril. 2023;119(1):1–2. https://doi.org/10.1016/j.fertnstert.2022.10.036.; Barcroft J., Dayoub N., Thong K.J. Fifteen year follow-up of embryos cryopreserved in cancer patients for fertility preservation. J Assist Reprod Genet. 2013;30(11):1407–13. https://doi.org/10.1007/s10815-013-0024-z.; Oktay K., Turan V., Bedoschi G. et al. Fertility preservation success subsequent to concurrent aromatase inhibitor treatment and ovarian stimulation in women with breast cancer. J Clin Oncol. 2015;33(22):2424–9. https://doi.org/10.1200/JCO.2014.59.3723.; Go K.J., Romanski P.A., Bortoletto P. et al. Meeting the challenge of unclaimed cryopreserved embryos. Fertil Steril. 2023;119(1):15–20. https://doi.org/10.1016/j.fertnstert.2022.09.323.; Pomeroy K.O., Comizzoli P., Rushing J.S. et al. The ART of cryopreservation and its changing landscape. Fertil Steril. 2022;117(3):469–76. https://doi.org/10.1016/j.fertnstert.2022.01.018.; Herraiz S., Buigues A., Díaz-García C. et al. Fertility rescue and ovarian follicle growth promotion by bone marrow stem cell infusion. Fertil Steril. 2018;109(5):908–918.e2. https://doi.org/10.1016/j.fertnstert.2018.01.004.; Umer A., Khan N., Greene D.L. et al. The therapeutic potential of human umbilical cord derived mesenchymal stem cells for the treatment of premature ovarian failure. Stem Cell Rev Rep. 2023;19(3):651–66. https://doi.org/10.1007/s12015-022-10493-y.; Del-Pozo-Lérida S., Salvador C., Martínez-Soler F. et al. Preservation of fertility in patients with cancer (Review). Oncol Rep. 2019;41(5):2607–14. https://doi.org/10.3892/or.2019.7063.; Peng X., Cheng C., Zhang X. et al. Design and application strategies of natural polymer Biomaterials in artificial ovaries. Ann Biomed Eng. 2023;51(3):461–78. https://doi.org/10.1007/s10439-022-03125-6.; Wu T., Gao Y.Y., Su J. et al. Three-dimensional bioprinting of artificial ovaries by an extrusion-based method using gelatin-methacryloyl bioink. Climacteric. 2022;25(2):170–8. https://doi.org/10.1080/13697137.2021.1921726.; Pei W., Fu L., Guo W. et al. Efficacy and safety of mesenchymal stem cell therapy for ovarian ageing in a mouse model. Stem Cell Res Ther. 2024;15(1):96. https://doi.org/10.1186/s13287-024-03698-0.; Jones G., Hughes J., Mahmoodi N. et al. What factors hinder the decision-making process for women with cancer and contemplating fertility preservation treatment? Hum Reprod Update. 2017;23(4):433–57. https://doi.org/10.1093/humupd/dmx009.; Barjasteh S., Farnam F., Elsous A. et al. Overcoming reproductive and psychological concerns of breast cancer survivors: a randomized controlled Trial. J Family Reprod Health. 2022;16(1):52–60. https://doi.org/10.18502/jfrh.v16i1.8594.; Logan S., Anazodo A. The psychological importance of fertility preservation counseling and support for cancer patients. Acta Obstet Gynecol Scand. 2019;98(5):583–97. https://doi.org/10.1111/aogs.13562.; Ko J.K.Y., Cheung C.S.Y., Cheng H.H.Y. et al. Knowledge, attitudes and intention on fertility preservation among breast cancer patients. Sci Rep. 2023;13(1):9645. https://doi.org/10.1038/s41598-023-36377-w.; Петров И.А., Дмитриева М.Л., Тихоновская О.А. и др. Современный взгляд на естественную фертильность. Российский вестник акушера-гинеколога. 2017;17(2):4–12. https://doi.org/10.17116/rosakush20171724-12.; https://www.gynecology.su/jour/article/view/2387
-
2
-
3Academic Journal
Authors: S. A. Kuleva, D. N. Kurochkina, D. A. Zvyagintseva, S. V. Ivanova, V. V. Tsyrlina, С. А. Кулева, Д. Н. Курочкина, Д. А. Звягинцева, С. В. Иванова, Е. В. Цырлина
Source: Meditsinskiy sovet = Medical Council; № 6 (2019); 176-183 ; Медицинский Совет; № 6 (2019); 176-183 ; 2658-5790 ; 2079-701X ; 10.21518/2079-701X-2019-6
Subject Terms: лучевая терапия, women, Hodgkin’s lymphoma, gonadotoxicity, fertility, alkylating drugs, radiation therapy, женщины, лимфома Ходжкина, гонадотоксичность, фертильность, алкилирующие препараты
File Description: application/pdf
Relation: https://www.med-sovet.pro/jour/article/view/2996/2925; Ansell S.M. Hodgkin lymphoma: 2016 update on diagnosis, risk-stratification, and management. Am. J. Hematol. 2016;4(91):434–442.; Smith R.S., Chen Q., Hudson M.M., Link M.P., Kun L., Weinstein H. et al. Prognostic Factors for Children With Hodgkin’s Disease Treated With Combined-Modality Therapy. J. Clin. Oncol. 2003;10(21):2026–2033.; Vecchi V., Pileri S., Burnelli R., Bontempi N., Comelli A., Testi A.M. et al. Treatment of pediatric Hodgkin disease tailored to stage, mediastinal mass, and age an Italian (AIEOP) Multicenter Study on 215 patients. Cancer. 1993;6(72):2049–2057.; Weiner M.A., Leventhal B.G., Marcus R., Brecher M., Ternberg J., Behm F.G. et al. Intensive chemotherapy and low-dose radiotherapy for the treatment of advanced-stage Hodgkin’s disease in pediatric patients: a Pediatric Oncology Group study. J. Clin. Oncol. 1991;9(9):1591–1598.; Kelly K.M., Sposto R., Hutchinson R., Massey V., McCarten K., Perkins S. et al. BEACOPP chemotherapy is a highly effective regimen in children and adolescents with high-risk Hodgkin lymphoma: a report from the Children’s Oncology Group. Blood. 2011;9(117):2596–603.; Tebbi C.K., Mendenhall N., London W.B., Williams J.L., de Alarcon P.A., Chauvenet A.R. Treatment of stage I, IIA, IIIA1 pediatric Hodgkin disease with doxorubicin, bleomycin, vincristine and etoposide (DBVE) and radiation: A Pediatric Oncology Group (POG) study. Pediatr. Blood Cancer. 2006;2(46):198–202.; Schellong G., Pötter R., Brämswig J., Wagner W., Prott F.J., Dörffel W. High cure rates and reduced long-term toxicity in pediatric Hodgkin’s disease: the German-Austrian multicenter trial DAL-HD-90. The German-Austrian Pediatric Hodgkin’s Disease Study Group. J. Clin. Oncol. 1999;12(17):3736–3744.; Кулева С.А., Анишкин М.Ю., Колыгин Б.А. Лимфома Ходжкина у детей и подростков: эффективность риск-адаптированного протокола СПбЛХ-05. Детская онкология. 2007;3-4:16.; Винокуров А.А. Лимфома Ходжкина и проблемы репродукции у мужчин. Клин. онкогематол. 2013;3:258–273.; Mauz-Körholz C., Metzger M.L., Kelly K.M., Schwartz C.L., Castellanos M.E., Dieckmann K. et al. Pediatric Hodgkin Lymphoma. J. Clin. Oncol. 2015;27(33):2975–2985.; Mauz-Körholz C., Hasenclever D., Dörffel W., Ruschke K., Pelz T., Voigt A. et al. Procarbazine-free OEPA-COPDAC chemotherapy in boys and standard OPPA-COPP in girls have comparable effectiveness in pediatric Hodgkin’s lymphoma: the GPOH-HD-2002 study. J. Clin. Oncol. 2010;23(28):3680–3686.; Behringer K., Breuer K., Reineke T., May M., Nogova L., Klimm B. Secondary Amenorrhea After Hodgkin’s Lymphoma Is Influenced by Age at Treatment, Stage of Disease, Chemotherapy Regimen, and the Use of Oral Contraceptives During Therapy: A Report From the German Hodgkin’s Lymphoma Study Group. J. Clin. Oncol. 2005;30(23):7555–7564.; Petraglia F., Hartmann B., Luisi S., Florio P., Kirchengast S., Santuz M. et al. Low levels of serum inhibin A and inhibin B in women with hypergonadotropic amenorrhea and evidence of high levels of activin A in women with hypothalamic amenorrhea. Fertil. Steril. 1998;5(70):907–912.; Yamoto M., Imai M., Otani H., Nakano R. Serum levels of inhibin A and inhibin B in women with normal and abnormal luteal function. Obstet. Gynecol. Clin. North. Am. 1997;89(5):773–776.; Папуша Л.И., Балашов Д.Н., Тюльпаков А.Н., Скоробогатова Е.В., Курникова Е.Е., Скворцова Ю.В. и др. Половое развитие и функция гонад у детей и подростков после аллогенной трансплантации гемопоэтических стволовых клеток с использованием различных режимов кондиционирования. Онкогематология. 2009;2:61–69.; Vet A., Laven J.S., de Jong F.H., Themmen A.P., Fauser B.C. Antimüllerian hormone serum levels: a putative marker for ovarian aging. Fertil. Steril. 2002;2(77):357–362.; Brämswig J.H., Riepenhausen M., Schellong G. Parenthood in adult female survivors treated for Hodgkin’s lymphoma during childhood and adolescence: a prospective, longitudinal study. Lancet Oncol. 2015;6(16):667–675.; Chow E.J., Stratton K.L., Leisenring W.M., Oeffinger K.C., Sklar C.A., Donaldson S.S. et al. Pregnancy after chemotherapy in male and female survivors of childhood cancer treated between 1970 and 1999: a report from the Childhood Cancer Survivor Study cohort. Lancet Oncol. 2016;5(17):567–76.; Oktem O., Kim S.S., Selek U., Schatmann G., Urman B. Ovarian and Uterine Functions in Female Survivors of Childhood Cancers. Oncologist. 2018;2(23):214–224.; Meirow D., Dor J., Kaufman B., Shrim A., Rabinovici J., Schiff E. et al. Cortical fibrosis and blood-vessels damage in human ovaries exposed to chemotherapy. Potential mechanisms of ovarian injury. Hum. Reprod. 2007;6(22):1626–1633.; Kalich-Philosoph L., Roness H., Carmely A., Fishel-Bartal M., Ligumsky H., Paglin S. et al. Cyclophosphamide triggers follicle activation and burnout prevents follicle loss and preserves fertility. Sci. Transl. Med. 2013;5(185):185ra62.; Himelstein-Braw R., Peters H., Faber M. Morphological study of the ovaries of leukaemic children. Br. J. Cancer. 1978;38(1):82-87.; Thomas-Teinturier C., Allodji R.S., Svetlova E., Frey M.A., Oberlin O., Millischer A.E. et al. Ovarian reserve after treatment with alkylating agents during childhood. Hum. Reprod. 2015;6(30):1437–1446.; Adriaens I., Smitz J., Jacquet P. The current knowledge on radiosensitivity of ovarian follicle development stages. Hum. Reprod. Update. 2009;3(15):359–377.; Wallace W.H., Thomson A.B., Saran F., Kelsey T.W. Predicting age of ovarian failure after radiation to a field that includes the ovaries. Int. J. Radiat. Oncol. Biol. Phys. 2005;3(62):738–744.; Hawkins M.M., Smith R.A. Pregnancy outcomes in childhood cancer survivors: Probable effects of abdominal irradiation. Int. J. Cancer. 1989;3(43):399–402.; Larsen E.C., Schmiegelow K., Rechnitzer C., Loft A., Müller J., Andersen A.N. Radiotherapy at a young age reduces uterine volume of childhood cancer survivors. Acta. Obstet. Gynecol. Scand. 2004;1(83):96–102.; Knopman J.M., Papadopoulos E.B., Grifo J.A., Fino M.E., Noyes N. Surviving childhood and reproductive-age malignancy: effects on fertility and future parenthood. Lancet Oncol. 2010;5(11):490–498.; Teh W.T., Stern C., Chander S., Hickey M. The Impact of Uterine Radiation on Subsequent Fertility and Pregnancy Outcomes. Biomed Res Int. 2014;2014:1–8.; Wallace W.H., Shalet S.M., Crowne E.C., Morris-Jones P.H., Gattamaneni H.R. Ovarian failure following abdominal irradiation in childhood: Natural history and prognosis. Clin. Oncol. 1989;2(1):75–79.; Chemaitilly W., Sklar C.A. Endocrine complications in long-term survivors of childhood cancers. Endocr. Relat. Cancer. 2010;3(17):141–159.; Sklar C.A., Mertens A.C., Mitby P., Whitton J., Stovall M., Kasper C. Premature Menopause in Survivors of Childhood Cancer: A Report From the Childhood Cancer Survivor Study. J. Natl. Cancer Inst. 2006;13(98):890–896.; Beek R.D., van den Heuvel-Eibrink M.M., Laven J.S., de Jong F.H., Themmen A.P., Hakvoort-Cammel F.G. et al. Anti-Müllerian Hormone Is a Sensitive Serum Marker for Gonadal Function in Women Treated for Hodgkin’s Lymphoma during Childhood. J. Clin. Endocr. Metab. 2007;10(92):3869–3874.; Johannsdottir I.M.R., Hamre H., Fosså S.D., Loge J.H., Drolsum L., Lund M.B. et al. Adverse Health Outcomes and Associations with Self-Reported General Health in Childhood Lymphoma Survivors. J. Adolesc. Young Adult Oncol. 2017;6(3):470-476.; Rach A.M., Crabtree V.M., Brinkman T.M., Zeltzer L., Marchak J.G., Srivastava D. Predictors of fatigue and poor sleep in adult survivors of childhood Hodgkin’s lymphoma: a report from the Childhood Cancer Survivor Study. J. Cancer Surviv. 2017;2(11):256–263.; Mastrо L., Ceppi M., Poggio F., Bighin C., Peccatori F., Demeestere I. et al. Gonadotropinreleasing hormone analogues for the prevention of chemotherapy-induced premature ovarian failure in cancer women: Systematic review and meta-analysis of randomized trials. Cancer Treat. Rev. 2014;5(40):675–683.; Behringer K., Wildt L., Mueller H., Mattle V., Ganitis P., van den Hoonaard B. et al. No protection of the ovarian follicle pool with the use of GnRH-analogues or oral contraceptives in young women treated with escalated BEACOPP for advanced-stage Hodgkin lymphoma. Final results of a phase II trial from the German Hodgkin Study Group. Ann. Oncol. 2010;10(21):2052–2060.; Elgindy E.A., El-Haieg D.O., Khorshid O.M., Ismail E.I., Abdelgawad M., Sallam H.N. et al. Gonadatrophin suppression to prevent chemotherapy-induced ovarian damage: a randomized controlled trial. Obstet. Gynecol. Clin. North. Am. 2013;1(121):78–86.; Peccatori F.A., Azim H.A. Jr., Orecchia R., Hoekstra H.J., Pavlidis N., Kesic V. et al. Cancer, pregnancy and fertility: ESMO Clinical Practice Guidelines for diagnosis, treatment and followup. Ann. Oncol. 2013;6(24):160-170.; Loren A.W., Mangu P.B., Beck L.N., Brennan L., Magdalinski A.J., Partridge A.H. et al. Fertility preservation for patients with cancer: American Society of Clinical Oncology clinical practice guideline update. J. Clin. Oncol. 2013;19(31):2500–2510.; Michaeli J., Weintraub M., Gross E., Ginosar Y., Ravitsky V., Eizenman E et al. Fertility Preservation in Girls. Int. J. Gynaecol. Obstet. 2012;2012:1–10.; Revel A., Revel-Vilk S., Aizenman E., Porat-Katz A., Safran A., Ben-Meir A. et al. At what age can human oocytes be obtained? Fertil. Steril. 2009;2(92):458–463.; Meirow D., Hardan I., Dor J., Fridman E., Elizur S., Ra’anani H., Slyusarevsky E. et al. Searching for evidence of disease and malignant cell contamination in ovarian tissue stored from hematologic cancer patients. Hum. Reprod. 2008;5(23):1007–1013.; Hovatta O. Methods for cryopreservation of human ovarian tissue. Reprod. Biomed. Online. 2005;6(10):729–734.; Donnez J., Dolmans M.M., Demylle D., Jadoul P., Pirard C., Squifflet J., Martinez-Madrid B. et al. Livebirth after orthotopic transplantation of cryopreserved ovarian tissue. Lancet. 2004;9443(364):1405–1410.; Лапина Е.Н., Лапина Е.Н., Быстрова О.В., Калугина А.С., Лисянская А.С., Татищева Ю.А. и др. Первая беременность в России после трансплантации криоконсервированной ткани яичника пациентке с лимфомой Ходжкина (описание случая). Проблемы репродукции. 2015;3(21):63.; Sanders J.E., Buckner C.D., Amos D., Levy W., Appelbaum F.R., Doney K. et al. Ovarian function following marrow transplantation for aplastic anemia or leukemia. J. Clin. Oncol. 1988;5(6):813–818.; Matsumoto M., Shinohara O., Ishiguro H., Shimizu T., Hattori K., Ichikawa M. et al. Ovarian function after bone marrow transplantation performed before menarche. Arch. Dis. Child. Educ. Pract. Ed. 1999;5(80):452–454.; Trueblood H.W., Enright L.P., Ray G.R., Kaplan H.S., Nelsen T.S. Preservation of ovarian function in pelvic radiation for Hodgkin’s disease. Arch. Surg. 1970;100(3):236–237.; Fernandez-Pineda I., Davidoff A.M., Lu L., Rao B.N., Wilson C.L., Srivastava D.K. et al. Impact of ovarian transposition before pelvic irradiation on ovarian function among long-term survivors of childhood Hodgkin lymphoma: A report from the St. Jude Lifetime Cohort Study. Pediatr. Blood Cancer. 2018;e27232.; Oktem O., Oktay О. Preservation of Menstrual Function in Adolescent and Young Females. Ann. N. Y. Acad. Sci. 2008;1(1135):237–243.; Gabriëlle G.J., Broekmans F.J., Dorland M., Habbema J.D., Looman C.W., te Velde E.R. Antral follicle counts by transvaginal ultrasonography are related to age in women with proven natural fertility. Fertil. Steril. 1999;5(72):845–851.; Crofton P.M., Thomson A.B., Evans A.E., Groome N.P., Bath L.E., Kelnar C.J. Is inhibin B a potential marker of gonadotoxicity in prepubertal children treated for cancer? Clinical endocrinology. 2003;3(58):296–301.
-
4Academic Journal
Authors: S. A. Kuleva, D. N. Kurochkina, D. A. Zvyagintseva, S. V. Ivanova, Ye. V. Tsyrlina, С. А. Кулева, Д. Н. Курочкина, Д. А. Звягинцева, С. В. Иванова, Е. В. Цырлина
Source: Meditsinskiy sovet = Medical Council; № 10 (2019); 164-171 ; Медицинский Совет; № 10 (2019); 164-171 ; 2658-5790 ; 2079-701X ; 10.21518/2079-701X-2019-10
Subject Terms: лучевая терапия, Hodgkin’s lymphoma, gonadotoxicity, fertility, alkylating drugs, radiation therapy, лимфома Ходжкина, гонадотоксичность, фертильность, алкилирующие препараты
File Description: application/pdf
Relation: https://www.med-sovet.pro/jour/article/view/3068/2992; Мерабишвили В.М. Выживаемость онкологических больных. СПб.: Коста, 2006. 440 с. [Merabishvili V.M. Survival rate of oncological patients. St. Petersburg: Costa, 2006. 440 p.] (In Russ.); Мень Т.Х., Поляков В.Г., Алиев М.Д. Эпидемиология злокачественных новообразований у детей в России. Онкопедиатрия. 2014;1:7-12. [Men’ T.H., Polyakov V.G., Aliev M.D. Epidemiology of malignant neoplasms in children in Russia. Onkopediatriya. 2014;1:7-12.] (In Russ.); Ларина Ю.В., Миненко С.В., Биячуев Э.Р., Пшонкин А.В., Хуажева Н.К., Птушкин В.В. Лечение распространенных форм лимфомы Ходжкина у подростков и молодых взрослых. Проблема эффективности и токсичности. Онкогематология. 2014;1:11-18. [Larina Yu.V., Minenko S.V., Biyachuev E.R., Pshonkin A.V., Khuazheva N.K., Ptushkin V.V. Treatment of common forms of Hodgkin lymphoma in adolescents and young adults. Problem of efficacy and toxicity. Onkogematologiya. 2014;1:11-18.] (In Russ.); Демина Е.А. Лимфома Ходжкина. Возможности современной терапии первичных больных. РМЖ. 2004;19:1113. [Demina E.A. Hodgkin’s lymphoma. Possibilities of modern therapy of primary patients. RMZh. 2004;19:1113.] (In Russ.); Кулева С.А., Колыгин Б.А. Отдаленные последствия противоопухолевой терапии у детей. Вопр. oнкол. 2012;4:454-463. [Kuleva S.A., Kolygin B.A. Remote consequences of antitumor therapy in children. Vopr. onkol. 2012;4:454-463.] (In Russ.); Колыгин Б.А., Кулева С.А. Последствия противоопухолевой терапии у детей. СПб., 2011. 184 с. [Kolygin B.A., Kuleva S.A. Consequences of antitumor therapy in children. St. Petersburg, 2011. 184 p.] (In Russ.); Демина Е.А. Лимфома Ходжкина: от Томаса Ходжкина до наших дней. Клин. oнкогематол., 2008;2:114-118. [Demina E.A. Hodgkin’s lymphoma: from Thomas Hodgkin to the present day. Klin. onkogematol., 2008;2:114-118.] (In Russ.); Pusey W.A. Cases of sarcoma and of Hodgkin’s disease treated by exposures to X-rays: a preliminary report. JAMA. 1902;38:166-169.; Peters V. A study of survival in Hodgkin’s disease treated by irradiation. Am. J. Roent. 1950;63:299-311.; De Vita V. Combination chemotherapy in the treatment of advanced Hodgkin’s disease. Ann. Intern. Med. 1970;73:881-895.; Bonadonna G. Combination chemotherapy of Hodgkin’s disease with adriamycin, bleomycin, vinblastine, and imidazole carboxamide versus MOPP. Cancer. 1975;36:252-259.; Prosnitz L.R. Consolidation radiotherapy in the treatment of advanced Hodgkin’s disease: is it dead? Int. J. Radiat. Oncol. Biol. Phys. 2003;56:605-608.; Винокуров А.А., Варфоломеева С.Р., Тарусин Д.И. Гонадотоксичность терапии лимфомы Ходжкина у подростков и молодых мужчин: актуальность проблемы и пути решения (обзор литературы). Онкогематология. 2011;2:12-19. [Vinokurov A.A., Varfolomeeva S.R., Tarusin D.I. Gonadotoxicity of Hodgkin’s lymphoma therapy in teenagers and young men: relevance of the problem and solutions (literature review). Onkogematologiya. 2011;2:12-19.] (In Russ.); Кулева С.А., Анишкин М.Ю., Колыгин Б.А. Сравнительный анализ двух риск-адаптированных программ, используемых в терапии лимфомы Ходжкина у детей и подростков. Вопр. oнкол. 2008;1:53-58. [Kuleva S.A., Anishkin M.Yu., Kolygin B.A. Comparative analysis of two risk-adapted programs used in therapy of Hodgkin’s lymphoma in children and adolescents. Vopr. onkol. 2008;1:53-58.] (In Russ.); Ansell S.M. Hodgkin lymphoma: 2016 update on diagnosis, risk-stratification, and management. Am. J. Hematol. 2016;91(4):434-442.; Smith R.S. Prognostic factors for children with Hodgkin’s disease treated with combinedmodality therapy. J. Clin. Oncol. 2003;21(10):2026-2033.; Weiner M.A. Intensive chemotherapy and lowdose radiotherapy for the treatment of advanced-stage Hodgkin’s disease in pediatric patients: a Pediatric Oncology Group Study. J. Clin. Oncol. 1991;9(9):1591-1598.; Vecchi V. Treatment of pediatric Hodgkin’s disease tailored to stage, mediastinal mass, and age. An Italian (AIEOP) multicenter study on 215 patients. Cancer. 1993;72(6):2049-2057.; Schellong G. High cure rates and reduced longterm toxicity in pediatric Hodgkin’s disease: the German-Austrian Multicenter Trial DAL-HD-90. J. Clin. Oncol. 1999;17(12):3736-3744.; Tebbi C.K., Mendenhall N., London W.B., Williams J.L., de Alarcon P.A., Chauvenet A.R. Treatment of stage I, IIA, IIIA1 pediatric Hodgkin disease with doxorubicin, bleomycin, vincristine and etoposide (DBVE) and radiation: a Pediatric Oncology Group (POG) study. Pediatr. Blood Cancer. 2006;46(2):198-202.; Kelly K.M., Sposto R., Hutchinson R., Massey V., McCarten K., Perkins S. et al. BEACOPP chemotherapy is a highly effective regimen in children and adolescents with high-risk Hodgkin lymphoma: a report from the Children’s Oncology Group. Blood. 2011;117(9):2596-2603.; Tebbi C.K., Mendenhall N.P., London W.B., Williams J.L., Hutchison R.E., Fitzgerald T.J. et al. Response-dependent and reduced treatment in lower risk Hodgkin lymphoma in children and adolescents, results of P9426: a report from the Children’s Oncology Group. Pediatr. Blood Cancer. 2012;59(7):1259-1265.; Кулева С.А., Анишкин М.Ю., Колыгин Б.А. Лимфома Ходжкина у детей и подростков: эффективность риск-адаптированного протоко- ла СПбЛХ-05. Детская онкология. 2007;3-4:16. [Kuleva S.A., Anishkin M.Yu., Kolygin B.A. Hodgkin’s lymphoma in children and teenagers: effectiveness of the risk-adapted protocol SPbHL- 05. Detskaya onkologiya. 2007;3-4:16.] (In Russ.); Mauz-Körholz C., Metzger M.L., Kelly K.M., Schwartz C.L., Castellanos M.E., Dieckmann K. et al. Pediatric Hodgkin Lymphoma. J. Clin. Oncol. 2015;33(27):2975-2985.; Mauz-Körholz C., Hasenclever D., Dörffel W., Ruschke K., Pelz T., Voigt A. et al. Procarbazinefree OEPA-COPDAC chemotherapy in boys and standard OPPA-COPP in girls have comparable effectiveness in pediatric Hodgkin’s lymphoma: the GPOH-HD-2002 study. J. Clin. Oncol. 2010;28(23):3680-3686.; Wyns C., Curaba M., Vanabelle B., Van Langendonckt A., Donnez J. Options for fertility preservation in prepubertal boys. Hum. Reprod. Update. 2010;16(3):312-328.; Lee S.H., Shin C.H. Reduced male fertility in childhood cancer survivors. Ann. Pediatr. Endocrinol. Metab. 2013;18(4):168-172. 28. Kaaij M.A., Echten-Arends J., Simons A.H., Kluin- Nelemans H.C. Fertility preservation after chemotherapy for Hodgkin lymphoma. Hematol. Oncol. 2010;28(4):168-179.; da Cunha M.F., Meistrich M.L., Fuller L.M., Cundiff J.H., Hagemeister F.B., Velasquez W.S. et al. Recovery of spermatogenesis after treatment for Hodgkin’s disease: limiting dose of MOPP chemotherapy. Clin. Oncol. 1984;2(6):571-577.; Jacob A., Barker H., Goodman A., Holmes J. Recovery of spermatogenesis following bone marrow transplantation. Bone Marrow Transplant, 1998;22(3):277-279.; Meistrich M.L. Effects of chemotherapy and radiotherapy on spermatogenesis in humans. Fertil. Steril. 2013;100(5):1180-1186.; Dohle G.R. Male infertility in cancer patients: Review of the literature. Int. J. Urol. 2010;17(4):327-331.; Tal R., Botchan A., Hauser R., Yogev L., Paz G., Yavetz H. Follow-up of sperm concentration and motility in patients with lymphoma. Hum. Reprod. 2000;15(9):1985-1988.; Kinsella T.J. Effects of radiation therapy and chemotherapy on testicular function. Prog. Clin. Biol. Res. 1989;302:157-171.; Shalet S.M., Tsatsoulis A., Whitehead E., Read G. Vulnerability of the human Leydig cell to radiation damage is dependent upon age. J. Endocrinol. 1989;120(1):161-165.; Apperley J.F., Reddy N. Mechanism and management of treatment-related gonadal failure in recipients of high dose chemoradiotherapy. Blood Rev. 1995;9(2):93-116.; Green D.M., Kawashima T., Stovall M. Fertility of male survivors of childhood cancer: a report from the childhood cancer survivor study. J. Clin. Oncol. 2010;28(2):332–339.; Howell S.J., Shalet S.M. Spermatogenesis after cancer treatment: damage and recovery. J. Natl. Cancer Inst. Monogr. 2005;34:12-17.; Белогурова М.Б., Диникина Ю.В., Лисянская А.С. Осложнения противоопухолевой терапии со стороны репродуктивной функции в результате лечения онкологических заболеваний у мальчиков. Онкопедиатрия. 2014;4:40-48. [Belogurova M.B., Dinikina Yu.V., Lisyanskaya A.S. Complications of antitumor therapy from the side of reproductive function as a result of treatment of oncological diseases in boys. Onkopediatriya. 2014;4:40-48.] (In Russ.); Oberfield S.E., Soranno D., Nirenberg A., Heller G., Allen J.C., David R. et al. Age at onset of puberty following high-dose central nervous system radiation therapy. Arch. Pediatr. Adolesc. Med. 1996;150(6):589-592.; Eichenauer D.A., Engert A. The evolving role of targeted drugs in the treatment of Hodgkin lymphoma. Expert Rev. Hematol. 2017;10(9):775-782.; Johannsdottir I.M., Karlstad O., Loge J.H., Fosså S.D., Kiserud C., Skurtveit S. Prescriptions of Antidepressants to Survivors of Cancer in Childhood, Adolescence, and Young Adulthood: A Population-Based Study. J. Adolesc. Young Adult Oncol. 2017;6(1):120-126.; Rach A.M., Crabtree V.M., Brinkman T.M., Zeltzer L., Marchak J.G., Srivastava D. et al. Predictors of fatigue and poor sleep in adult survivors of childhood Hodgkin’s lymphoma: a report from the Childhood Cancer Survivor Study. J. Cancer Surviv. 2017;11(2):256-263.; Hallak J., Mahran A.M., Agarwal A. Characteristics of cryopreserved semen from men with lymphoma. J. Assist. Reprod. Genet. 2000;17(10):591-594.; Marmor D., Elefant E., Dauchez C., Roux C. Semen analysis in Hodgkin’s disease before the onset of treatment. Cancer. 1986;57(10):1986-1987.; Padron O.F., Sharma R.K., Thomas A.J., Agarwal A. Effects of cancer on spermatozoa quality after cryopreservation: a 12-year experience. Fertil. Steril. 1997;67(2):326-331.; Paoli D., Gallo M., Rizzo F., Spanò M., Leter G., Lombardo F. et al. Testicular cancer and sperm DNA damage: short- and long-term effects of antineoplastic treatment. Andrology. 2015;3(1):122-128.; Schmidt K.T., Andersen C.Y. ISFP Practice Committee. Recommendations for fertility preservation in patients with lymphomas. Assist. Reprod. Genet. 2012;29(6):473-477.; Anderson R.A., Weddell A., Spoudeas H.A., Douglas C., Shalet S.M., Levitt G. et al. Do doctors discuss fertility issues before they treat young patients with cancer? Hum. Reprod. 2008;23(10):2246-2251.; Bahadur G. Cryobiology ethics of human reproduction. Semin. Reprod. Med. 2002;20(1):75-83.; Hovav Y., Dan-Goor M., Yaffe H., Almagor M. Electroejaculation before chemotherapy in adolescents and young men with cancer. Fertil. Steril. 2001;75(4):811-813.; Lee S.J., Schover L.R., Partridge A.H., Patrizio P., Wallace W.H., Hagerty K. et al. American Society of Clinical Oncology. American Society of Clinical Oncology recommendations on fertility preservation in cancer patients. J. Clin. Oncol. 2006;24(18):2917-2931.; Frederickx V., Michiels A., Goossens E., De Block G., Van Steirteghem A.C., Tournaye H. Recovery, survival and functional evaluation by transplantation of frozen-thawed mouse germ cells. Hum. Reprod. 2004;19(4):948-953.; Nieschlag E. Clinical trials in male hormonal contraception. Contraception. 2010;82(5):457-470.; Винокуров А.А., Варфоломеева С.Р., Тарусин Д.И., Абдуллаев Р.Т., Гретцов Е.М., Луценко И.Н. и др. Фертильность подростков и молодых мужчин, излеченных от лимфомы Ходжкина. Злокачественные опухоли. 2012;1:18-27. [Vinokurov A.A., Varfolomeeva S.R., Tarusin D.I., Abdullaev R.T., Grettsov E.M., Lutsenko I.N. et al. Fertility of teenagers and young men cured from Hodgkin’s lymphoma. Zlokachestvennye opuholi. 2012;1:18-27.] (In Russ.); Krawczuk-Rybak M., Solarz E., Wysocka J., Matysiak M., Gadomski A., Kazanowska B. et al. Testicular function after treatment for acute lymphoblastic leukemia (all) in prepubertal and pubertal boys. Pediatr. Hematol. Oncol. 2009;26(7):504-514.
-
5Academic Journal
Authors: M. B. Belogurova, Yu. V. Dinikina, A. S. Lisyanskaya, N. I. Tapilskaya, G. G. Radulesku, T. D. Viktorovich, L. I. Shats, E. D. Chavpetsova, М. Б. Белогурова, Ю. В. Диникина, А. С. Лисянская, Н. И. Тапильская, Г. Г. Радулеску, Т. Д. Викторович, Л. И. Шац, Э. Д. Чавпецова
Source: Russian Journal of Pediatric Hematology and Oncology; Том 2, № 4 (2015); 34-41 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 2, № 4 (2015); 34-41 ; 2413-5496 ; 2311-1267 ; 10.17650/2311-1267-2015-2-4
Subject Terms: гормонотерапия, adolescents, pediatric oncology, menorrhagia, fertility, chemotherapy, mielosuprescia, reproduction, gonadal toxicity, hormonotherapy, подростки, детская онкология, меноррагия, фертильность, химиотерапия, миелосупрессия, репродукция, гонадотоксичность
File Description: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/157/152; Brenner H., Coebergh J.W., Parkin D.M. et al. Up-to-date monitoring of childhood cancer long-term survival in Europe: Leukaemias and lymphomas. Ann Oncol 2007;18(9):1569–77.; Blumenfeld Z., von Wolff M. GnRHanalogues and oral contraceptives for fertility preservation in women during chemotherapy. Hum Reprod Update 2008;14(6):543–52.; Michaeli J., Weintraub M., Gross E. et al. Fertility preservation in girls. Obstet Gynecol Int 2012;2012:139193.; Del Mastro L., Ceppi M., Poggio F. et al. Gonadotropin-releasing hormone analogues for the prevention of chemotherapy-induced premature ovarian failure in cancer women: Systematic review and meta-analysis of randomized trials. Cancer Treat Rev 2014;40(5):675–83.; Белогурова М.Б. Диникина Ю.В., Тапильская Н.И. и др. Нарушение фертильности как осложнение лечения онкологических заболеваний в детском и подростковом возрасте (обзор литературы). Евразийский онкологический журнал 2014;1:77–86. [Belogurova М.B., Dinikina Yu.V., Tapilskaya N.I. Violation of fertility as a complication of cancer treatment in children and adolescents (review). Yevraziyskiy onkologicheskiy zhurnal = Eurasian Journal of Oncology 2014;1:77–86. (In Russ.)].; Brown J., Demetri G. Challenges in the development of platelet growth factors: low expectations for low counts. Curr Hematol Rep 2002;1(2):110–8.; Meirow D., Rabinovici J., Katz D. et al. Prevention of severe menorrhagia in oncology patients with treatment-induced thrombocytopenia by luteinizing hormonereleasing hormone agonist and depomedroxyprogesterone acetate. Cancer 2006;107(7):1634–41.; Committee opinion no. 606: Options for prevention and management of heavy menstrual bleeding in adolescent patients undergoing cancer treatment. Obstet Gynecol 2014;124(2 Pt 1):397–402.; Hanker J.P. Gastrointestinal disease and oral contraception. Am J Obstet Gynecol 1990;163(6 Pt 2):2204–7.; Bates J.S., Buie L.W., Woodis C.B. Management of menorrhagia associated with chemotherapy-induced thrombocytopenia in women with hematologic malignancy. Pharmacotherapy 2011;31(11):1092–110.; Kline R., Fennewald L., Vore M. et al. Oral contraceptives a cause of hyperbilirubinemia in stem cell transplant patients. J Pediatr Hematol Oncol 1999;21(5):436–40.; Quaas A.M., Ginsburg E.S. Prevention and treatment of uterine bleeding in hematologic malignancy. Eur J Obstet Gynecol Reprod Biol 2007;134(1):3–8.; Loren A.S., Mangu P.B., Beck L.N. et al. Fertility preservation for patients with cancer: American Society of Clinical Oncology clinical practice guideline update. J Clin Oncol 2013;31(19):2500–10.; Patridge A.H., Gelber S., Peppercorn J. et al. Web-based survey of fertility issues in young women with breast cancer. J Clin Oncol 2004;22(20):4174–83.; Диникина Ю.В. Оптимизация методов сохранения фертильности у девочек с онкологической патологией. Диссертация на соискание ученой степени к.м.н. СПб.: [б.н.], 2011. 145 с. [Dinikina Yu.V. Optimization methods of fertility preservation for girls with cancer pathology. Dissertation for the degree of Ph.D. SPb.: [unnumbered], 2011. 145 p. (In Russ.)].; Peccatori F.A., Azim H.A. Jr, Orrecchia R. et al.; ESMO Guidelines Working Group. Cancer, pregnancy and fertility: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2013;24 Suppl 6:vi160–70.; Sklar C.A., Mertens A.C., Mitby P. et al. Premature menopause in survivors of childhood cancer: a report from the childhood cancer survivor study. J Natl Cancer Inst 2006;98(13):890–6.; Ortin T., Shostak C., Donaldson S. Gonadal status and reproductive function following treatment for Hodgkin's disease in childhood: the Stanford experience. Int J Radiat Oncol Biol Phys 1990;19(4):873–80.; Beck-Fruchter R., Weiss A., Shalev E. GnRH agonist therapy as ovarian protectants in female patients undergoing chemotherapy: a review of the clinical data. Hum Reprod Update 2008;14(6):553–61.; Demeestere I., Brice P., Peccatori F. et al. Gonadotropin-releasing hormone agonist for the prevention of chemotherapy-induced ovarian failure in patients with lymphoma: 1-year follow-up of a prospective randomized trial. J Clin Oncol 2013;31(7):903–9.; Blumenfeld Z. How to preserve fertility in young women exposed to chemotherapy? The role of GnRH agonist cotreatment in addition to cryopreservation of embrya, oocytes, or ovaries. Oncologist 2007;12(9):1044–54.; Clowse M.E., Behera M.A., Anders C.K. et al. Ovarian preservation by GnRH agonists during chemotherapy: a meta-analysis. J Womens Health (Larchmt) 2009;18(3):311–9.; Chen H., Li J., Cui T., Hu L. Adjuvant gonadotropin-releasing hormone analogues for the prevention of chemotherapy induced premature ovarian failure in premenopausal women. Cochrane Database Syst Rev 2011;(11):CD008018.; Slater C.A., Liang M.H., McCune J.W. et al. Preserving ovarian function in patients receiving cyclophosphamide. Lupus 1999;8(1):3–10.; Ataya M., McKanna J., Weintraub A. et al. A luteinizing hormone-releasing hormone agonist for the prevention of chemotherapy-induced ovarian follicular loss in rats. Cancer Res 1985;45(8):3651–6.; Bokser L., Szende B., Schally A. Protective effects of D-Trp6-luteinising hormone-releasing hormone microcapsules against cyclophosphamide-induced gonadotoxicity in female rats. Br J Cancer 1990;61(6):861–5.; Ataya K., Ramahi-Ataya A. Reproductive performance of female rats treated with cyclophosphamide and/or LHRH agonist. Reprod Toxicol 1993;7(3):229–35.; Ataya K., Rao L.V., Lawrence E., Kimmel R. Luteinizing hormone-releasing hormone agonist inhibits cyclophosphamideinduced ovarian follicular depletion in rhesus monkeys. Biol Reprod 1995;52(2):365–72.; Imai A., Sugiyama M., Furui T. et al. Direct protection by gonadotropin-releasing hormone analog from doxorubicin-induced granulosa cell damage. Gynecol Obstet Invest 2007;63(2):102–6.; Letterie G.S. Anovulation in the prevention of cytotoxic-induced follicular attrition and ovarian failure. Hum Reprod 2004;19(4):831–7.; Blumenfeld Z. Gender difference: fertility preservation in young women but not in men exposed to gonadotoxic chemotherapy. Minerva Endocrinol 2007;32(1):23–34.; Lobo R.A. Potential options for preservation fertility in women. N Engl J Med 2005;353(1):64–73.; Oktay K., Sönmezer M., Oktem O. et al. Absence of conclusive evidence for the safety and efficacy of gonadotropin-releasing hormone analogue treatment in protecting against chemotherapy-induced gonadal injury. Oncologist 2007;12(9):1055–66.; Sönmezer M., Oktay K. Fertility preservation in female patients. Hum Reprod Update 2004;10(3):251–66.; Kitajima Y., Endo T., Nagasawa K. et al. Hyperstimulation and a gonadotropinreleasing; hormone agonist modulate ovarian vascular permeability by altering expression of the tight junction protein claudin-5. Endocrinology 2006;147(2):694–9.; Yu N., Chan C., Tang O. et al. Effect of pituitary downregulation on antral follicle count, ovarian volume and stromal blood flow measured by three-dimensional ultrasound with power Doppler prior to ovarian stimulation. Hum Reprod 2004;19(12):2811–5.; Blumenfeld Z. Gynecologic concerns for young women exposed to gonadotoxic chemotherapy. Curr Opin Obstet Gynecol 2003;15(5):359–70.; Janssens R., Brus L., Cahill D. et al. Direct ovarian effects and safety aspects of GnRH agonists and antagonists. Hum Reprod Update 2000;6(5):505–18.; Kolesnick R.N., Krönke M. Regulation of ceramide production and apoptosis. Annu Rev Physiol 1998;60:643–65.; Tilly J.L. Commuting the death sentence: how oocytes strive to survive. Nat Rev Mol Cell Biol 2001;2(11):838–48.; Johnson J., Canning J., Kaneko T. et al. Germline stem cells and follicular renewal in; the postnatal mammalian ovary. Nature 2004;428(6979):145–50.; Johansen J., Riis B., Hassager C. et al. The effect of a gonadotropin-releasing hormone agonist analog (nafarelin) on bone metabolism. J Clin Endocrinol Metab 1988;67(4):701–6.; Fogelman I. Gonadotropin-releasing hormone agonists and the skeleton. Fertil Steril 1992;57(4):715–24.; Scialli A., Jestila K., Simon J. Leuprolide acetate and bone mineral density measured by quantitive radiography. Fertil Steril 1993;59(3):674–6.; Qublan H., Amarin Z., Tahat Y. et al. Ovarian cyst formation following Gnrh agonist administration in IVF cycles: incidence and impact. Hum Reprod 2006;21(3):640–4.; von Wolff M., Kämmerer U., Kollmann Z. et al. Combination of gonadotropin-releasing hormone (GnRH) agonists with GnRH antagonists before chemotherapy reduce but does not; completely prevent a follicle-stimulating hormone flare-up. Fertil Steril 2011;95(1):452–4.; Blumenfeld Z., Avivi I., Eckman A. et al. Gonadotropin-releasing hormone agonist decreases chemotherapy-induced gonadotoxicity and premature ovarian failure in young female patients with Hodgkin lymphoma. Fertil Steril 2008;89(1):166–73.; Banzal A., Patel F., Rai B. et al. Gonadotrophin releasing hormone analogues for ovarian function preservation in young females undergoing chemotherapy. Asian Pac J Cancer Prev 2014;15(5):2185–90.; Somers E.C., Marder W., Christman G.M. et al. Use of a gonadotropin-releasing hormone analog for protection against premature ovarian failure during cyclophosphamide therapy in women with severe lupus. Arthritis Rheum 2005;52(9):2761–7.; Practice Committee of the American Society for Reproductive Medicine. Testing and interpreting measures of ovarian reserve: a committee opinion. Fertil Steril 2012;98(6):1407–15.; https://journal.nodgo.org/jour/article/view/157
-
6Academic Journal
Authors: A. A. Vinokurov, G. A. Novichkova, A. G. Rumyantsev, А. А. Винокуров, Г. А. Новичкова, А. Г. Румянцев
Source: Russian Journal of Pediatric Hematology and Oncology; Том 2, № 4 (2015); 42-50 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 2, № 4 (2015); 42-50 ; 2413-5496 ; 2311-1267 ; 10.17650/2311-1267-2015-2-4
Subject Terms: лучевая терапия, chemotherapy, sperm cryopreservation, oncofertility, gonadal toxicity, adolescent and young adult oncology, Hodgkin's lymphoma, acute lymphoblastic leukemia, testicular cancer, alkylating agents, spermatogenesis, cytostatics, infertility after chemotherapy, infertility in adolescent, radiation therapy, химиотерапия, криоконсервация спермы, онкофертильность, гонадотоксичность, онкология у подростков и молодых взрослых, лимфома Ходжкина, острый лимфобластный лейкоз, опухоль яичка, алкилирующие препараты, сперматогенез, цитостатики, бесплодие после химиотерапии, бесплодие у подростков
File Description: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/158/153; Hirsch M., Lunenfeld B., Modan M. et al. Spermarche the age of onset of sperm emission. J Adolesc Health Care 1985;6(1):35–9.; Nielsen C.T., Skakkebaek N.E., Richardson D.W. et al. Onset of the release of spermatozoa (spermarche) in boys in relation to age, testicular growth, pubic hair, and height. J Clin Endocrinol Metab 1986;62(3):532–5.; Nieschlag E., Behre H. Andrology. Male Reproductive Health and Dysfunction. 3 ed. Springer-Verlag Berlin Heidelberg, 2010. Pp. 16–17.; Zhengwei Y., McLachlan R., Bremner W., Wreford N. Quantitative (stereological) study of the normal spermatogenesis in the adult monkey (Macaca fascicularis). J Androl 1997;18(6):681–7.; Винокуров А.А. Лимфома Ходжкина и проблемы репродукции у мужчин. Клиническая онкогематология 2013;6(3):258–73. [Vinokurov A.A. Hodgkin’s lymphoma and male fertility disorders. Klinicheskaya onkogematologiya = Clinical oncohematology 2013;6(3):258–73. (In Russ.)].; Винокуров А.А., Варфоломеева С.Р., Тарусин Д.И., Моисеева Т.Н. Оценка гонадотоксичности терапии по схеме ВЕАСОРР-14 у молодых мужчин, излеченных от лимфомы Ходжкина. Клиническая онкогематология 2011;4(3):235–9. [Vinokurov A.A., Varfolomeeva S.R., Tarusin D.I., Moiseeva T.N. Assessment of gonadal toxicity of Hodgkin's lymphoma in young males treated by BEACOPP-14 protocol. Klinicheskaya onkogematologiya = Clinical oncohematology 2011;4(3):235–9. (In Russ.)].; Steger K., Rey R., Louis F. et al. Reversion of the differentiated phenotype and maturation block in Sertoli cells in pathological human testis. Hum Reprod 1999;14(1):136–43.; von Eckardstein S., Simoni M., Bergmann M. et al. Serum inhibin B in combination with serum follicle-stimulating hormone (FSH) is a more sensitive marker than serum FSH alone for impaired spermatogenesis in men, but cannot predict the presence of sperm in testicular tissue samples. J Clin Endocrinol Metab 1999;84(7):2496–501.; Chung K., Irani J., Knee G. et al. Sperm cryopreservation for male patients with cancer: an epidemiological analysis at the University of Pennsylvania. Eur J Obstet Gynecol Reprod Biol 2004;113 Suppl 1:S7–11.; Williams D., Karpman E., Sander J. et al. Pretreatment semen parameters in men with cancer. J Urol 2009;181(2):736–40.; Marmor D., Elefant E., Dauchez C., Roux C. Semen analysis in Hodgkin's disease before the onset of treatment. Cancer 1986;57(10):1986–7.; Padron O.F., Sharma R.K., Thomas A.J. Jr, Agarwal A. Effects of cancer on spermatozoa quality after cryopreservation: a 12-year experience. Fertil Steril 1997;67(2):326–31.; Viviani S., Ragni G., Santoro A. et al. Testicular dysfunction in Hodgkin's disease before and after treatment. Eur J Cancer 1991;27(11):1389–92.; Bahadur G., Ozturk O., Muneer A. et al. Semen quality before and after gonadotoxic treatment. Hum Reprod 2005;20(3):774–81.; Gandini L., Lombardo F., Salacone P. et al. Testicular cancer and Hodgkin's disease: evaluation of semen quality. Hum Reprod 2003;18(4):796–801.; Rofeim O., Gilbert B. Normal semen parameters in cancer patients presenting for cryopreservation before gonadotoxic therapy. Fertil Steril 2004;82(2):505–6.; Rueffer U., Breuer K., Josting A. et al. Male gonadal dysfunction in patients with Hodgkin's disease prior to treatment. Ann Oncol 2001;12(9):1307–11.; Buch J.P., Kolon T.F., Maulik N. et al. Cytokines stimulate lipid membrane peroxidation of human sperm. Fertil Steril 1994;62(1):186–8.; Dousset B., Hussenet F., Daudin M. et al. Seminal cytokine concentrations (IL-1beta, IL-2, IL-6, sR IL-2, sR IL-6), semen parameters and blood hormonal status in male infertility. Hum Reprod 1997;12(7):1476–9.; Fedder J., Ellerman-Eriksen S. Effect of cytokines on sperm motility and ionophorestimulated acrosome reaction. Arch Androl 1995;35(3):173–85.; Meistrich M.L. Male gonadal toxicity. Pediatr Blood Cancer 2009;53(2):261–6.; Spitz S. The histological effects of nitrogen mustards on human tumors and tissues. Cancer 1948;1(3):383–98.; Seshadri T., Seymour J.F., McArthur G.A. Oligospermia in a patient receiving imatinib therapy for the hypereosinophilic syndrome. N Engl J Med 2004;351(20):2134–5.; Mariani S., Basciani S., Fabbri A. et al. Severe oligozoospermia in a young man with chronic myeloid leukemia on long-term treatment with imatinib started before puberty. Fertil Steril 2011;95(3):1120.e15–7.; Trottmann M., Becker A.J., Stadler T. et al. Semen quality in men with malignant diseases before and after therapy and the role of cryopreservation. Eur Urol 2007;52(2):355–67.; Meistrich M.L., Vassilopoulou-Sellin R., Lipshultz L.I. Adverse effects of treatment: gonadal dysfunction. 7 ed. Philadelphia: Lippincott Williams & Wilkins, 2005.; Colpi G.M., Contalbi G.F., Nerva F. et al. Testicular function following chemoradiotherapy. Eur J Obstet Gynecol Reprod Biol 2004;113 Suppl 1:S2–S6.; Meistrich M.L., Finch M., da Cunha M.F. et al. Damaging effects of fourteen chemotherapeutic drugs on mouse testis cells. Cancer Res 1982;42(1):122–31.; Chatterjee R., Haines G.A., Perera D.M. et al. Testicular and sperm DNA damage after treatment with fludarabine for chronic lymphocytic leukaemia. Hum Reprod 2000;15(4):762–6.; Abu-Baker S.O. Gemcitabine impacts histological structure of mice testis and embryonic organs. Pak J Biol Sci 2009;12(8):607–15.; Savkovic N., Green S., Pecevski J., Maric N. The effect of mitomycin on the fertility and the induction of meiotic chromosome rearrangements in mice and their first generation progeny. Can J Genet Cytol 1977;19(3):387–93.; Heim C., Minniear K., Dann C. Imatinib has deleterious effects on differentiating spermatogonia while sparing spermatogonial stem cell self renewal. Reprod Toxicol 2011;31(4):454–63.; Heikens J., Behrendt H., Adriaanse R., Berghout A. Irreversible gonadal damage in male survivors of pediatric Hodgkin's disease. Cancer 1996;78(9):2020–4.; Shafford E.A., Kingston J.E., Malpas J.S. et al. Testicular function following the treatment of Hodgkin's disease in childhood. Br J Cancer 1993;68(6):1199–204.; Aubier F., Flamant F., Brauner R. et al. Male gonadal function after chemotherapy for solid tumors in childhood. J Clin Oncol 1989;7(3):304–9.; Ben Arush M.W., Solt I., Lightman A. et al. Male gonadal function in survivors of childhood Hodgkin and non-Hodgkin lymphoma. Pediatr Hematol Oncol 2000;17(3):239–45.; Dhabhar B.N., Malhotra H., Joseph R. et al. Gonadal function in prepubertal boys following treatment for Hodgkin's disease. Am J Pediatr Hematol Oncol 1993;15(3):306–10.; Nurmio M., Keros V., Lähteenmäki P. et al. Effect of childhood acute lymphoblastic leukemia therapy on spermatogonia populations and future fertility. J Clin Endocrinol Metab 2009;94(6):2119–22.; Meistrich M.L. Impact of cyclophosphamide on long-term reduction in sperm count in men treated with combination chemotherapy for Ewing and soft tissue sarcomas. Cancer 1992;70(11):2703–12.; Reinmuth S., Hohmann C., Rendtorff R. et al. Impact of chemotherapy and radiotherapy in childhood on fertility in adulthood: the FeCt-survey of childhood cancer survivors in Germany. J Cancer Res Clin Oncol 2013;139(12):2071–8.; Sherins R.J., Olweny C.L., Ziegler J.L. Gynecomastia and gonadal dysfunction in adolescent boys treated with combination chemotherapy for Hodgkin's disease. N Engl J Med 1978;299(1):12–6.; Green D.M., Brecher M.L., Lindsay A.N. et al. Gonadal function in pediatric patients following treatment for Hodgkin disease. Med Pediatr Oncol 1981;9(3):235–44.; Puscheck E., Philip P.A., Jeyendran R.S. Male fertility preservation and cancer treatment. Cancer Treat Rev 2004;30(2):173–80.; Kinsella T.J., Trivette G., Rowland J. et al. Long-term follow-up of testicular function following radiation therapy for early-stage Hodgkin's disease. J Clin Oncol 1989;7(6):718–24.; Giwercman A., von der Maase H., Berthelsen J.G. et al. Localized irradiation of testes with carcinoma in situ: effects on Leydig cell function and eradication of malignant germ cells in 20 patients. J Clin Endocrinol Metab 1991;73(3):596–603.; Hahn E.W., Feingold S.M., Nisce L. Aspermia and recovery of spermatogenesis in cancer patients following incidental gonadal irradiation during treatment: a progress report. Radiology 1976;119(1):223–5.; Socie G., Salooja N., Cohen A. et al. Nonmalignant late effects after allogeneic stem cell transplantation. Blood 2003;101(9):3373–85.; Howell S.J., Shalet S.M. Spermatogenesis after cancer treatment: damage and recovery. J Natl Cancer Inst Monogr 2005;(34):12–7.; Meistrich M.L., Shetty G. Inhibition of spermatogonial differentiation by testosterone. J Androl 2003;24(2):135–48.; Meistrich M.L., Shetty G. Hormonal suppression for fertility preservation in males and females. Reproduction 2008;136(6):691–701.; Feldschuh J., Brassel J., Durso N., Levine A. Successful sperm storage for 28 years. Fertil Steril 2005;84(4):1017.; Horne G., Atkinson A.D., Pease E.H. et al. Live birth with sperm cryopreserved for 21 years prior to cancer treatment: case report. Hum Reprod 2004;19(6):1448–9.; Clarke G.N., Liu D.Y., Baker H.W. Recovery of human sperm motility and ability to interact with the human zona pellucida after more than 28 years of storage in liquid nitrogen. Fertil Steril 2006;86(3):721–2.; Agarwal A., Ranganathan P., Kattal N. et al. Fertility after cancer: a prospective review of assisted reproductive outcome with banked semen specimens. Fertil Steril 2004;81(2):342–8.; Saito K., Suzuki K., Iwasaki A. et al. Sperm cryopreservation before cancer chemotherapy helps in the emotional battle against cancer. Cancer 2005;104(3):521–4.; Schover L.R., Brey K., Lichtin A. et al. Knowledge and experience regarding cancer, infertility, and sperm banking in younger male survivors. J Clin Oncol 2002;20(7):1880–9.; Janssens P.M., Beerendonk C.C., Blokzijl E. et al. Cryopreservation of semen of adolescents and young adult men with cancer. Ned Tijdschr Geneeskd 2004;148(40):1981–4.; Blatt J. Pregnancy outcome in long-term survivors of childhood cancer. Med Pediatr Oncol 1999;33(1):29–33.; Byrne J., Rasmussen S.A., Steinhorn S.C. et al. Genetic disease in offspring of long-term survivors of childhood and adolescent cancer. Am J Hum Genet 1998;62(1):45–52.; Dodds L., Marrett L.D., Tomkins D.J. et al. Case-control study of congenital anomalies in children of cancer patients. BMJ 1993;307(6897):164–8.; Green D.M., Whitton J.A., Stovall M. et al. Pregnancy outcome of partners of male survivors of childhood cancer: a report from the Childhood Cancer Survivor Study. J Clin Oncol 2003;21(4):716–21.; Kenney L.B., Nicholson H.S., Brasseux C. et al. Birth defects in offspring of adult survivors of childhood acute lymphoblastic leukemia. A Childrens Cancer Group/ National Institutes of Health Report. Cancer 1996;78(1):169–76.; Meistrich M.L., Byrne J. Genetic disease in offspring of long-term survivors of childhood and adolescent cancer treated with potentially mutagenic therapies. Am J Hum Genet 2002;70(4):1069–71.; Senturia Y.D., Peckham C.S. Children fathered by men treated with chemotherapy for testicular cancer. Eur J Cancer 1990;26(4):429–32.; Signorello L.B., Mulvihill J.J., Green D.M. et al. Congenital anomalies in the children of cancer survivors: a report from the childhood cancer survivor study. J Clin Oncol 2012;30(3):239–45.; Папуша Л.И., Младова Е.С., Хилькевич Л.В. и др. Сохранение фертильности у пациенток, перенесших трансплантацию гемопоэтических стволовых клеток. Гематология и трансфузиология 2012;57(3):70. [Papusha L.I., Mladova E.S., Khilkevich L.V. et al. Preservation of fertility in of patients after hematopoietic stem cell transplantation. Gematologiya i transfuziologiya = Hematology and Transfusiology 2012;57(3):70. (In Russ.)].; Поварова А.А., Гаврилов В.М., Младова Е.С. и др. Возможности ВРТ у пациенток с онкологическими заболеваниями. Международная научно-практическая конференция «Репродуктивные технологии в онкологии – 2015». Сборник материалов конференции. С. 35. [Povarova A.A., Gavrilov V.M., Mladova E.S. et al. Features of assisted reproductive technology in patients with cancer. International scientific-practical conference ‟Reproductive Technologies in Oncology – 2015”. Conference materials. P. 35. (In Russ.)].; Винокуров А.А., Варфоломеева С.Р., Тарусин Д.И. и др. Фертильность подростков и молодых мужчин, излеченных от лимфомы Ходжкина. Гематология и трансфузиология 2013;58(2):11–8. [Vinokurov A.A., Varfolomeeva S.R., Tarusin D.I. et al. Fertility of adolescents and young men treated for Hodgkin’s lymphoma. Gematologiya i transfuziologiya = Hematology and Transfusiology 2013;58(2):11–8. (In Russ.)].; https://journal.nodgo.org/jour/article/view/158
-
7Academic Journal
Authors: А. А. Винокуров, С. Р. Варфоломеева, Д. В. Тарусин, Р. Т. Абдуллаев, Е. М. Гретцов, И. Н. Луценко, А. В. Пивник, Т. Н. Моисеева
Source: Malignant tumours; Том 2, № 1 (2012); 18-28 ; Злокачественные опухоли; Том 2, № 1 (2012); 18-28 ; 2587-6813 ; 2224-5057
Subject Terms: бесплодие, ВЕАСОРР-14, гонадотоксичность
File Description: application/pdf
Relation: https://www.malignanttumors.org/jour/article/view/60/64; Armitage J.O. Long-term toxicity of the treatment of Hodgkin’s disease. Ann Oncol 1998;9(Suppl 5):133–6.; Puscheck E., Philip P.A., Jeyendran R.S. Male fertility preservation and cancer treatment. Cancer Treat Rev 2004 Apr;30(2):173–80; Vogel E.W., Nivard M.J. International Commission for Protection Against Environmental Mutagens and Carcinogens. The subtlety of alkylating agents in reactions with biological macromolecules. Mutat Res 1994 Feb 1;305(1):13–32.; Vogel E.W., Barbin A., Nivard M.J., Bartsch H. Nucleophilic selectivity of alkylating agents and their hypermutability in Drosophila as predictors of carcinogenic potency in rodents. Carcinogenesis 1990 Dec;11(12):2211–7.}; Ehrenberg L. Covalent binding of genotoxicagents to proteins and nucleic acids. IARC Sci Pub 1984;(59):107–14.; Angerer J., Ewers U., Wilhelm M. Human biomonitoring: state of the art. Int J Hyg Environ Health 2007 May;210(3–4):201–28.; Krawczuk-Rybak M., Solarz E., Wojtkowska M., Wysocka J., Matysiak M., Gadomski A., Kazanowska B., Sega-Pondel D., Gonadal function in young men after the treatment for Hodgkin lymphoma. Pediatr Endocrinol Diabetes Metab. 2009;15(2):85-92.; Swerdlov A.J., Barber J.A., Hudson G.V. et al. J Clin Oncol 2000;18(3):498–508.; Волкова М.А., Клиническая онкогематология: Руководство для врачей, 2-е изд., Москва «Медицина» 2007,1120, стр. 699-700 10. Johnson J.E., Boone W.R., Blackhurst D.W. Manual versus computer-automated semen analyses. Part II. Determination of the working range of a computer-automated semen analyzer. HamiltonThorne Research Technical Note 2217 (July, 2001).; Афифи А, Эйзен С. Статистический анализ: Подход с использованием ЭВМ. - М.:Мир, 1982; 488.; Справочник по прикладной статистике. В 2-х т. Т.1: Пер. с англ./ Под ред. Э.Ллойда, У.Ледермана, Ю.Н.Тюрина.- М.: Финансы и статистики, 1989; - 510с. Т. 2: 1990; - 526с.; Леонов В.П. Обработка экспериментальных данных на программируемых микрокалькуляторах. Томск, изд-во ТГУ. - 1990. - 376с.; Кендалл М., Стьюарт А. Статистические выводы и связи. Пер. с англ. М.: Главная редакция физ.-мат. литературы. 1973, с. 899.; WHO laboratory manual for examination of human semen and sperm-cervical mucus interaction / пер. с англ. Р.А.Нерсеяна под научн. ред. рус. перевода Л.Ф. Курило — 4-е издание. — М.: Изд-во «МедПресс», 2001. — 144 с.; van der Kaaij M.A., Heutte N., van Echten-Arends J., Raemaekers J.M., Carde P. et.al. Sperm quality before treatment in patients with early stage Hodgkin›s lymphoma enrolled in EORTC-GELA Lymphoma Group trials. 2009 Dec;94(12):1691-7. Epub 2009 Oct 22.; Rueffer U., Вreuer K., Josting A. et.al. Male gonadal dysfunction in patients with Hodgkin disease prior to treatment. Ann of Oncol. Vol 12 №9 Sept 2001 p1307-1313.; Bahadur G., Ozturk O., Muneer A., Wafa R. et.al. Hum Reprod. Semen quality before and after gonadotoxic treatment. 2005 Mar;20(3):774-81. Epub 2005 Feb 2.; Buch J.P., Colon T.F., Maulik N. et. al. Cytokines stimulate lipid membrane peroxidation of human sperm. Fertil Steril 1994. 62 186-8; Dousset B., Hussenet F., Daudin M. et. al. Seminal cytokine concentrations. ( I L - i p, 1L-2, IL-6, sR 1L2, sR IL-6), semen parameters and blood hormonal status in male infertility Hum Reprod 1997, 12 1476-9; Gruschwitz M.S., Brezinschek R., Brezinschek H.P. Cytokine levels in the seminal plasma of infertile males. J Androl 1996, 17’ 158-63; Huleihel M., Lunenfeld E. Levy A. et. al. Distinct expression levels of cytokines and soluble cytokine receptors in seminal plasma of fertile and infertile men Fertil Steril 1996, 66 135-9; Schulte H.M., Bamberger C.M., Elsen H. et. al. Systemic mterleukin-1 alpha and interleukin-2 secretion in response to acute stress and to corticotropin-releasing hormone in humans. Eur J Clin Invest 1994, 24 773-7; Shimonovitz S., Barak V., Zakut D. et. al. High concentration of soluble interleukin-2 receptors in ejaculate with low sperm motillty. Hum Reprod 1994. 9 653-5; Bahadur G., Gandini L., Lombardo F., Salacone P., Paoli D., Anselmo A.P., Culasso F., Dondero F. and Lenzi A. (2003) Testicular cancer and Hodgkin’s disease: evaluation of semen quality. Hum Reprod 18,796–801.; Sieniawski М., Reineke T., Nogova L., Diehl V. et al. Fertility in male patients with advanced HL treated with BEACOPP. Blood 2008 Jan 1;111(1):71–6.; Пивник А.В., Расстриин Н.А., Моисеева Т.Н. и соавт. Результаты лечения лимфогранулематоза по протоколу МОРР-АВVD в сочетании с лучевой терапией ( десятилетнее наблюдение). Тер. архив 2006,№ 8,57-62; Hamilton V.M., Norris C., Bunin N., Goldwein J.W., Bunin G.R., Lange B., Meadows A.T. Cyclophosphamidebased, seven-drug hybrid and low-dose involved field radiation for the treatment of childhood and adolescent Hodgkin disease. J Pediatr Hematol Oncol 2001;23(2):84–8.; Винокуров А. А., Варфоломеева С.Р., Тарусин Д.И. Гонадотоксичность терапии лимфомы Ходжкина у подростков и молодых мужчин: актуальность проблемы и пути решения (обзор литературы); Онкогематология. 2011г; № 2 - С.12-18.; https://www.malignanttumors.org/jour/article/view/60
-
8Academic Journal
Subject Terms: ЛИМФОМА ХОДЖКИНА, HODGKIN''S LYMPHOMA, ГОНАДОТОКСИЧНОСТЬ, ФЕРТИЛЬНОСТЬ, КРИОКОНСЕРВАЦИЯ СПЕРМЫ
File Description: text/html
-
9Academic Journal
Authors: Винокуров, А., Варфоломеева, С., Тарусин, Д., Моисеева, Т.
Subject Terms: ЛИМФОМА ХОДЖКИНА, ХИМИОТЕРАПИЯ, ВЕАСОРР-14, ГОНАДОТОКСИЧНОСТЬ, СТЕРИЛЬНОСТЬ, ВОССТАНОВЛЕНИЕ СПЕРМАТОГЕНЕЗА, КРИОКОНСЕРВАЦИЯ СПЕРМЫ, HODGKIN''S LYMPHOMA
File Description: text/html
-
10Academic Journal
Authors: Винокуров, А., Варфоломеева, С., Тарусин, Д.
Subject Terms: ЛИМФОМА ХОДЖКИНА, АЛКИЛИРУЮЩИЕ ПРЕПАРАТЫ, ГОНАДОТОКСИЧНОСТЬ, КРИОКОНСЕРВАЦИЯ СПЕРМЫ
File Description: text/html
-
11Academic Journal
Source: Гематология и трансфузиология.
Subject Terms: 03 medical and health sciences, 0302 clinical medicine, ЛИМФОМА ХОДЖКИНА, HODGKIN'S LYMPHOMA, ГОНАДОТОКСИЧНОСТЬ, ФЕРТИЛЬНОСТЬ, КРИОКОНСЕРВАЦИЯ СПЕРМЫ, 3. Good health
File Description: text/html
-
12Academic Journal
Source: Клиническая онкогематология. Фундаментальные исследования и клиническая практика.
Subject Terms: ЛИМФОМА ХОДЖКИНА, ХИМИОТЕРАПИЯ, ВЕАСОРР-14, ГОНАДОТОКСИЧНОСТЬ, СТЕРИЛЬНОСТЬ, ВОССТАНОВЛЕНИЕ СПЕРМАТОГЕНЕЗА, КРИОКОНСЕРВАЦИЯ СПЕРМЫ, HODGKIN'S LYMPHOMA, 3. Good health
File Description: text/html
-
13Academic Journal
Source: Онкогематология.
Subject Terms: 03 medical and health sciences, 0302 clinical medicine, ЛИМФОМА ХОДЖКИНА, АЛКИЛИРУЮЩИЕ ПРЕПАРАТЫ, ГОНАДОТОКСИЧНОСТЬ, КРИОКОНСЕРВАЦИЯ СПЕРМЫ, 3. Good health
File Description: text/html