-
1Academic Journal
Source: Медицина и организация здравоохранения, Vol 10, Iss 1 (2025)
Subject Terms: Medicine (General), R5-920, персонализированная медицина, молекулярно-генетическое тестирование, ДНК
-
2Academic Journal
Authors: K. K. Laktionov, V. V. Breder, A. K. Allakhverdiev, A. M. Kazakov, A. E. Gorokhov, P. V. Kononets, D. I. Yudin, D. L. Stroyakovsky, Ya. S. Akhmadiyarova, V. V. Kozlov, A. M. Fedun, T. F. Ibragimov, T. A. Sannikova, I. V. Plokhotenko, К. К. Лактионов, В. В. Бредер, А. К. Аллахвердиев, А. М. Казаков, А. Е. Горохов, П. В. Кононец, Д. И. Юдин, Д. Л. Строяковский, Я. С. Ахмадиярова, В. В. Козлов, А. М. Федунь, Т. Ф. Ибрагимов, Т. А. Санникова, И. В. Плохотенко
Contributors: The company Autonomous Non-Profit Organization National Society of Onco-Pulmonologists is the sponsor and coordinator of this study. The authors would like to thank AstraZeneca., Компания АНО «НООП» является спонсором и координатором данного исследования. Коллектив авторов выражает благодарность компании AstraZeneca.
Source: Meditsinskiy sovet = Medical Council; Online First ; Медицинский Совет; Online First ; 2658-5790 ; 2079-701X
Subject Terms: молекулярно-генетическое тестирование, surgical treatment, adjuvant therapy, molecular genetic testing, оперативное лечение, адъювантная терапия
File Description: application/pdf
Relation: https://www.med-sovet.pro/jour/article/view/8936/7770; Kratzer TB, Bandi P, Freedman ND, Smith RA, Travis WD, Jemal A, Siegel RL. Lung cancer statistics, 2023. Cancer. 2024;130(8):1330−1348. https://doi.org/10.1002/cncr.35128.; Каприн АД, Старинский ВВ, Шахзадовой АО (ред.). Злокачественные новообразования в России в 2021 году. М.: МНИОИ им. П.А. Герцена − филиал ФГБУ «НМИЦ радиологии» Минздрава России; 2022. 252 с. Режим доступа: https://oncology-association.ru/wp-content/uploads/2022/11/zlokachestvennye-novoobrazovaniya-v-rossii-v-2021-g_zabolevaemost-i-smertnost.pdf.; de Groot PM, Wu CC, Carter BW, Munden RF. The epidemiology of lung cancer. Transl Lung Cancer Res. 2018;7(3):220−233. https://doi.org/10.21037/tlcr.2018.05.06.; Garinet S, Wang P, Mansuet-Lupo A, Fournel L, Wislez M, Blons H. Updated Prognostic Factors in Localized NSCLC. Cancers. 2022;14(6):1400. https://doi.org/10.3390/cancers14061400.; Mueller M. ED08.01 Surgery of Early-Stage NSCLC. J Thorac Oncol. 2017;12(1):S40−S42. https://doi.org/10.1016/j.jtho.2016.11.038.; McDonald F, De Waele M, Hendriks LE, Faivre-Finn C, Dingemans AC, Van Schil PE. Management of stage I and II nonsmall cell lung cancer. Eur Respir J. 20173;49(1):1600764. https://doi.org/10.1183/13993003.00764-2016.; Raman V, Yang CJ, Deng JZ, D’Amico TA. Surgical treatment for early stage non-small cell lung cancer. J Thorac Dis. 2018;10(Suppl. 7):S898−S904. https://doi.org/10.21037/jtd.2018.01.172.; Veronesi G, Novellis P, Voulaz E, Alloisio M. Robot-assisted surgery for lung cancer: State of the art and perspectives. Lung Cancer. 2016;101:28−34. https://doi.org/10.1016/j.lungcan.2016.09.004.; Lackey A, Donington JS. Surgical management of lung cancer. Semin Intervent Radiol. 2013;30(2):133–140. https://doi.org/10.1055/s-0033-1342954.; Arriagada R, Bergman B, Dunant A, Le Chevalier T, Pignon JP, Vansteenkiste J. Cisplatin-based adjuvant chemotherapy in patients with completely resected non-small-cell lung cancer. N Engl J Med. 2004;350(4):351−360. https://doi.org/10.1056/NEJMoa031644.; Kim MH, Kim SH, Lee MK, Eom JS. Recent Advances in Adjuvant Therapy for Non-Small-Cell Lung Cancer. Tuberc Respir Dis. 2024;87(1):31−39. https://doi.org/10.4046/trd.2023.0085.; Pignon JP, Tribodet H, Scagliotti GV, Douillard JY, Shepherd FA, Stephens RJ et al. Lung adjuvant cisplatin evaluation: a pooled analysis by the LACE Collaborative Group. J Clin Oncol. 2008;26(21):3552−3559. https://doi.org/10.1200/JCO.2007.13.9030.; Rotolo F, Dunant A, Le Chevalier T, Pignon JP, Arriagada R. Adjuvant cisplatin-based chemotherapy in nonsmall-cell lung cancer: new insights into the effect on failure type via a multistate approach. Ann Oncol. 2014;25(11):2162–2166. https://doi.org/10.1093/annonc/mdu442.; Felip E, Altorki N, Zhou C, Csőszi T, Vynnychenko I, Goloborodko O et al. Adjuvant atezolizumab after adjuvant chemotherapy in resected stage IB-IIIA non-small-cell lung cancer (IMpower010): a randomised, multicentre, open-label, phase 3 trial. Lancet. 2021;398(10308):1344−1357. https://doi.org/10.1016/S0140-6736(21)02098-5.; Wakelee HA, Altorki NK, Zhou C, Csőszi T, Vynnychenko IO, Goloborodko O et al. IMpower010: Final disease-free survival (DFS) and second overall survival (OS) interim results after ≥5 years of follow up of a phase III study of adjuvant atezolizumab vs best supportive care in resected stage IB-IIIA non-small cell lung cancer (NSCLC). J Clin Oncol. 2024;42(17):LBA8035. https://doi.org/10.1200/JCO.2024.42.17_suppl.LBA803.; Cascone T, Awad MM, Spicer J, He J, Lu S, Sepesi B et al. LBA1 CheckMate 77T: Phase III study comparing neoadjuvant nivolumab (NIVO) plus chemotherapy (chemo) vs neoadjuvant placebo plus chemo followed by surgery and adjuvant NIVO or placebo for previously untreated, resectable stage II–IIIb NSCLC. Ann Oncol. 2023;34(Suppl. 2):S1295. https://doi.org/10.1016/j.annonc.2023.10.050.; Oselin K, Shim BY, Okada M, Bryl M, Bonanno L, Demirag G. Pembrolizumab vs placebo for early-stage non‒small-cell lung cancer after resection and adjuvant therapy: Subgroup analysis of patients who received adjuvant chemotherapy in the phase 3 PEARLS/ KEYNOTE-091 study. J Clin Oncol. 2023;41(16):8520. https://doi.org/10.1200/JCO.2023.41.16_suppl.8520.; Tsuboi M, Herbst RS, John T, Kato T, Majem M, Grohé C et al. Overall Survival with Osimertinib in Resected EGFR-Mutated NSCLC. N Engl J Med. 2023;389(2):137−147. https://doi.org/10.1056/NEJMoa2304594.; Wu YL, Dziadziuszko R, Ahn JS, Barlesi F, Nishio M, Lee DH et al. Alectinib in Resected ALK-Positive Non–Small-Cell Lung Cancer. N Engl J Med. 2024;390(14):1265–1276. https://doi.org/10.1056/NEJMoa2310532.; Kim MH, Kim SH, Lee MK, Eom JS. Recent Advances in Adjuvant Therapy for Non-Small-Cell Lung Cancer. Tuberc Respir Dis. 2024;87(1):31–39. https://doi.org/10.4046/trd.2023.0085.
-
3Academic Journal
Authors: K. K. Laktionov, K. A. Sarantseva, L. A. Nelyubina, S. V. Gamayunov, E. A. Kolesnikova, M. G. Gordiev, К. К. Лактионов, К. А. Саранцева, Л. А. Нелюбина, С. В. Гамаюнов, Е. А. Колесникова, М. Г. Гордиев
Source: Siberian journal of oncology; Том 23, № 2 (2024); 72-81 ; Сибирский онкологический журнал; Том 23, № 2 (2024); 72-81 ; 2312-3168 ; 1814-4861
Subject Terms: NGS, molecular genetic testing, KRAS, KRAS G12C, sotorasib, молекулярно-генетическое тестирование, соторасиб
File Description: application/pdf
Relation: https://www.siboncoj.ru/jour/article/view/3039/1217; Prior I.A., Hood F.E., Hartley J.L. The Frequency of Ras Mutations in Cancer. Cancer Res. 2020; 80(14): 2969–74. doi:10.1158/0008-5472.CAN-19-3682.; BiernackaA., Tsongalis P.D., PetersonJ.D., deAbreu F.B., BlackC.C., Gutmann E.J., Liu X., Tafe L.J., Amos C.I., Tsongalis G.J. The potential utility of re-mining results of somatic mutation testing: KRAS status in lung adenocarcinoma. Cancer Genet. 2016; 209(5): 195–8. doi:10.1016/j.cancergen.2016.03.001.; Ettinger D.S., Wood D.E., Aisner D.L., Akerley W., Bauman J.R., Bharat A., Bruno D.S., Chang J.Y., Chirieac L.R., D’Amico T.A., DeCamp M., Dilling T.J., Dowell J., Gettinger S., Grotz T.E., Gubens M.A., Hegde A., Lackner R.P., Lanuti M., Lin J., Loo B.W., Lovly C.M., Maldonado F., Massarelli E., Morgensztern D., Ng T., Otterson G.A., Pacheco J.M., Patel S.P., Riely G.J., Riess J., Schild S.E., Shapiro T.A., Singh A.P., Stevenson J., Tam A., Tanvetyanon T., Yanagawa J., Yang S.C., Yau E., Gregory K., Hughes M. Non-Small Cell Lung Cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2022; 20(5):497–530. doi:10.6004/jnccn.2022.0025.; Злокачественное новообразование бронхов и легкого. Клинические рекомендации. Министерство здравоохранения Российской Федерации. 2021.; NCCN Clinical Practice Guidelines in Oncology (NCCN Guide-lines®). Non-Small Cell Lung Cancer. Version 1.2024.; Kessler D., Gmachl M., Mantoulidis A., Martin L.J., Zoephel A., Mayer M., Gollner A., Covini D., Fischer S., Gerstberger T., Gmaschitz T., Goodwin C., Greb P., Häring D., Hela W., Hoffmann J., Karolyi-Oezguer J., Knesl P., Kornigg S., Koegl M., Kousek R., Lamarre L., Moser F., Munico-Martinez S., Peinsipp C., Phan J., Rinnenthal J., Sai J., Salamon C., Scherbantin Y., Schipany K., Schnitzer R., Schrenk A., Sharps B., Siszler G., Sun Q., Waterson A., Wolkerstorfer B., Zeeb M., Pearson M., Fesik S.W., McConnell D.B. Drugging an undruggable pocket on KRAS. Proc Natl Acad Sci U S A. 2019; 116(32): 15823–9. doi:10.1073/pnas.1904529116.; Malumbres M., Barbacid M. RAS oncogenes: the first 30 years. Nat Rev Cancer. 2003; 3(6): 459–65. doi:10.1038/nrc1097. Erratum in: Nat Rev Cancer. 2003; 3(9): 708.; Friedlaender A., Drilon A., Weiss G.J., Banna G.L., Addeo A. KRAS as a druggable target in NSCLC: Rising like a phoenix after decades of development failures. Cancer Treat Rev. 2020; 85. doi:10.1016/j.ctrv.2020.101978.; Ferrer I., Zugazagoitia J., Herbertz S., John W., Paz-Ares L., Schmid-Bindert G. KRAS-Mutant non-small cell lung cancer: From biology to therapy. Lung Cancer. 2018; 124: 53–64. doi:10.1016/j.lungcan.2018.07.013.; Román M., Baraibar I., López I., Nadal E., Rolfo C., Vicent S., Gil-Bazo I. KRAS oncogene in non-small cell lung cancer: clinical perspectives on the treatment of an old target. Mol Cancer. 2018; 17(1): 33. doi:10.1186/s12943-018-0789-x.; Poulin E.J., Bera A.K., Lu J., Lin Y.J., Strasser S.D., Paulo J.A., Huang T.Q., Morales C., Yan W., Cook J., Nowak J.A., Brubaker D.K., Joughin B.A., Johnson C.W., DeStefanis R.A., Ghazi P.C., Gondi S., Wales T.E., Iacob R.E., Bogdanova L., Gierut J.J., Li Y., Engen J.R., Perez-Mancera P.A., Braun B.S., Gygi S.P., Lauffenburger D.A., Westover K.D., Haigis K.M. Tissue-Specific OncogenicActivity of KRASA146T. Cancer Discov. 2019;9(6): 738–55. doi:10.1158/2159-8290.CD-18-1220.; Yuan T.L., Amzallag A., Bagni R., Yi M., Afghani S., Burgan W., Fer N., Strathern L.A., Powell K., Smith B., Waters A.M., Drubin D., Thomson T., Liao R., Greninger P., Stein G.T., Murchie E., Cortez E., Egan R.K., Procter L., Bess M., Cheng K.T., Lee C.S., Lee L.C., Fellmann C., Stephens R., Luo J., Lowe S.W., Benes C.H., McCormick F. Differential Effector Engagement by Oncogenic KRAS. Cell Rep. 2018; 22(7): 1889–902. doi:10.1016/j.celrep.2018.01.051.; Muñoz-Maldonado C., Zimmer Y., Medová M. A Comparative Analysis of Individual RAS Mutations in Cancer Biology. Front Oncol. 2019; 9: 1088. doi:10.3389/fonc.2019.01088.; Riely G.J., Kris M.G., Rosenbaum D., Marks J., Li A., Chitale D.A., Nafa K., Riedel E.R., Hsu M., Pao W., Miller V.A., Ladanyi M. Frequency and distinctive spectrum of KRAS mutations in never smokers with lung adenocarcinoma. Clin Cancer Res. 2008; 14(18): 5731–4. doi:10.1158/1078-0432.CCR-08-0646.; Slebos R.J., Hruban R.H., Dalesio O., Mooi W.J., Offerhaus G.J., Rodenhuis S. Relationship between K-ras oncogene activation and smoking in adenocarcinoma of the human lung. J Natl Cancer Inst. 1991; 83(14):1024–7. doi:10.1093/jnci/83.14.1024.; Cox A.D., Fesik S.W., Kimmelman A.C., Luo J., Der C.J. Drugging the undruggable RAS: Mission possible? Nat Rev Drug Discov. 2014;13(11): 828–51. doi:10.1038/nrd4389.; Ryan M.B., Corcoran R.B. Therapeutic strategies to target RAS-mutant cancers. Nat Rev Clin Oncol. 2018; 15(11): 709–20. doi:10.1038/s41571-018-0105-0.; Awad M.M., Gadgeel S.M., Borghaei H., Patnaik A., Yang J.C., Powell S.F., Gentzler R.D., Martins R.G., Stevenson J.P., Altan M., Jalal S.I., Panwalkar A., Gubens M., Sequist L.V., Saraf S., Zhao B., Piperdi B., Langer C.J. Long-Term Overall Survival From KEYNOTE-021 Cohort G: Pemetrexed and Carboplatin With or Without Pembrolizumab as First-Line Therapy for Advanced Nonsquamous NSCLC. J Thorac Oncol. 2021; 16(1): 162–8. doi:10.1016/j.jtho.2020.09.015.; Ostrem J.M., Peters U., Sos M.L., Wells J.A., Shokat K.M. K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature. 2013; 503(7477): 548–51. doi:10.1038/nature12796.; Lito P., Solomon M., Li L.S., Hansen R., Rosen N. Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism. Science. 2016; 351(6273): 604–8. doi:10.1126/science.aad6204.; Janes M.R., Zhang J., Li L.S., Hansen R., Peters U., Guo X., Chen Y., Babbar A., Firdaus S.J., Darjania L., Feng J., Chen J.H., Li S., Li S., Long Y.O., Thach C., Liu Y., Zarieh A., Ely T., Kucharski J.M., Kessler L.V., Wu T., Yu K., Wang Y., Yao Y., Deng X., Zarrinkar P.P., Brehmer D., Dhanak D., Lorenzi M.V., Hu-Lowe D., Patricelli M.P., Ren P., Liu Y. Targeting KRAS Mutant Cancers with a Covalent G12C-Specific Inhibitor. Cell. 2018; 172(3): 578–89. doi:10.1016/j.cell.2018.01.006.; Zeng M., Lu J., Li L., Feru F., Quan C., Gero T.W., Ficarro S.B., Xiong Y., Ambrogio C., Paranal R.M., Catalano M., Shao J., Wong K.K., Marto J.A., Fischer E.S., Jänne P.A., Scott D.A., Westover K.D., Gray N.S. Potent and Selective Covalent Quinazoline Inhibitors of KRAS G12C. Cell Chem Biol. 2017; 24(8): 1005–16. doi:10.1016/j.chembiol.2017.06.017.; Liu J., Kang R., Tang D. The KRAS-G12C inhibitor: activity and resistance. Cancer Gene Therapy. 2022; 29: 875–8. doi:10.1038/s41417-021-00383-9.; Dy G.K., Govindan R., Velcheti V., Falchook G.S., Italiano A., Wolf J., Sacher A.G., Takahashi T., Ramalingam S.S., Dooms C., Kim D.W., Addeo A., Desai J., Schuler M., Tomasini P., Hong D.S., Lito P., Tran Q., Jones S., Anderson A., Hindoyan A., Snyder W., Skoulidis F., Li B.T. Long-Term Outcomes and Molecular Correlates of Sotorasib Efficacy in Patients With Pretreated KRAS G12C-Mutated Non-Small-Cell Lung Cancer: 2-Year Analysis of CodeBreaK 100. J Clin Oncol. 2023; 41(18): 3311–7. doi:10.1200/JCO.22.02524.; Canon J., Rex K., SaikiA.Y., Mohr C., Cooke K., Bagal D., Gaida K., Holt T., Knutson C.G., Koppada N., Lanman B.A., Werner J., Rapaport A.S., San Miguel T., Ortiz R., Osgood T., Sun J.R., Zhu X., McCarter J.D., Volak L.P., Houk B.E., Fakih M.G., O’Neil B.H., Price T.J., Falchook G.S., Desai J., Kuo J., Govindan R., Hong D.S., Ouyang W., Henary H., Arvedson T., Cee V.J., Lipford J.R. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature. 2019; 575(7781): 217–23. doi:10.1038/s41586-019-1694-1.; Jänne P.A., Riely G.J., Gadgeel S.M., Heist R.S., Ou S.I., Pacheco J.M., Johnson M.L., Sabari J.K., Leventakos K., Yau E., Bazhenova L., Negrao M.V., Pennell N.A., Zhang J., Anderes K., Der-Torossian H., Kheoh T., Velastegui K., Yan X., Christensen J.G., Chao R.C., Spira A.I. Adagrasib in Non-Small-Cell Lung Cancer Harboring a KRASG12C Mutation. N Engl J Med. 2022; 387(2): 120–31. doi:10.1056/NEJ-Moa2204619.; Ou S.I., Jänne P.A., Leal T.A., Rybkin I.I., Sabari J.K., Barve M.A., Bazhenova L., Johnson M.L., Velastegui K.L., Cilliers C., Christensen J.G., Yan X., Chao R.C., Papadopoulos K.P. First-in-Human Phase I/IB Dose-Finding Study of Adagrasib (MRTX849) in Patients with Advanced KRASG12C Solid Tumors (KRYSTAL-1). J Clin Oncol. 2022; 40(23):2530–8. doi:10.1200/JCO.21.02752.; Skoulidis F., Li B.T., Dy G.K., Price T.J., Falchook G.S., Wolf J., Italiano A., Schuler M., Borghaei H., Barlesi F., Kato T., Curioni-Fonte-cedro A., Sacher A., Spira A., Ramalingam S.S., Takahashi T., Besse B., Anderson A., Ang A., Tran Q., Mather O., Henary H., Ngarmchamnanrith G., Friberg G., Velcheti V., Govindan R. Sotorasib for Lung Cancers with KRAS p.G12C Mutation. N Engl J Med. 2021; 384(25): 2371–81. doi:10.1056/NEJMoa2103695.; Hong D.S., Fakih M.G., Strickler J.H., Desai J., Durm G.A., Shapiro G.I., Falchook G.S., Price T.J., Sacher A., Denlinger C.S., Bang Y.J., Dy G.K., Krauss J.C., Kuboki Y., Kuo J.C., Coveler A.L., Park K., Kim T.W., Barlesi F., Munster P.N., Ramalingam S.S., Burns T.F., Meric-Bernstam F., Henary H., Ngang J., Ngarmchamnanrith G., Kim J., Houk B.E., Canon J., Lipford J.R., Friberg G., Lito P., Govindan R., Li B.T. KRASG12C Inhibition with Sotorasib in Advanced Solid Tumors. N Engl J Med. 2020;383(13): 1207–17. doi:10.1056/NEJMoa1917239.; Scheffler M., Ihle M.A., Hein R., Merkelbach-Bruse S., Scheel A.H., Siemanowski J., Brägelmann J., Kron A., Abedpour N., Ueckeroth F., Schüller M., Koleczko S., Michels S., Fassunke J., Pasternack H., Heydt C., Serke M., Fischer R., Schulte W., Gerigk U., Nogova L., Ko Y.D., Abdulla D.S.Y., Riedel R., Kambartel K.O., Lorenz J., Sauerland I., Randerath W., Kaminsky B., Hagmeyer L., Grohé C., Eisert A., Frank R., Gogl L., Schaepers C., Holzem A., Hellmich M., Thomas R.K., Peifer M., Sos M.L., Büttner R., Wolf J. K-ras Mutation Subtypes in NSCLC and Associated Co-occuring Mutations in Other Oncogenic Pathways. J Thorac Oncol. 2019; 14(4): 606–16. doi:10.1016/j.jtho.2018.12.013.; Dunnett-Kane V., Nicola P., Blackhall F., Lindsay C. Mechanisms of resistance to KRAS(G12C) inhibitors. Cancers. 2021; 13: 151.; https://www.siboncoj.ru/jour/article/view/3039
-
4Academic Journal
Authors: T. T. Tikhomirova, A. S. Tyulyandina, A. A. Rumyantsev, E. R. Israelyan, T. V. Kekeeva, O. V. Vedrova, M. L. Filipenko, L. N. Lyubchenko, I. A. Demidova, E. N. Imyanitov, S. Y. Andreev, S. V. Khokhlova, V. V. Saevets, G. B. Statsenko, L. A. Kolomiets, S. A. Tkachenko, I. A. Koroleva, A. S. Lisyanskaya, O. A. Bakashvili, L. I. Krikunova, E. P. Solovieva, D. M. Ponomarenko, L. Y. Vladimirova, S. E. Krasilnikov, V. B. Shirinkin, D. D. Sakaeva, E. A. Rumyantseva, S. A. Emeliyanov, D. L. Stroyakovskiy, E. G. Novikova, E. A. Ronina, V. I. Vladimirov, O. Y. Novikova, L. S. Zagumennova, V. V. Gorobtsova, E. V. Cherepanova, E. N. Pashkova, V. M. Moiseyenko, F. G. Ivanova, D. P. Udovitsa, V. V. Karaseva, S. A. Tyulyandin, Т. Е. Тихомирова, А. С. Тюляндина, А. А. Румянцев, Э. Р. Исраелян, Т. В. Кекеева, О. В. Ведрова, М. Л. Филипенко, Л. Н. Любченко, И. А. Демидова, Е. Н. Имянитов, С. Ю. Андреев, С. В. Хохлова, В. В. Саевец, Г. Б. Стаценко, Л. А. Коломиец, С. А. Ткаченко, И. А. Королева, А. С. Лисянская, О. А. Бакашвили, Л. И. Крикунова, Е. П. Соловьева, Д. М. Пономаренко, Л. Ю. Владимирова, С. Э. Красильников, В. Б. Ширинкин, Д. Д. Сакаева, Е. А. Румянцева, С. А. Емельянов, Д. Л. Строяковский, Е. Г. Новикова, Е. А. Ронина, В. И. Владимиров, О. Ю. Новикова, Л. С. Загуменнова, В. В. Горобцова, Е. В. Черепанова, Е. Н. Пашкова, В. М. Моисеенко, Ф. Г. Иванова, Д. П. Удовица, В. В. Карасева, С. А. Тюляндин
Source: Malignant tumours; Том 13, № 4 (2023); 46-59 ; Злокачественные опухоли; Том 13, № 4 (2023); 46-59 ; 2587-6813 ; 2224-5057
Subject Terms: молекулярно-генетическое тестирование, BRCA1 / 2 mutations, molecular genetic testing, мутации в генах BRCA1/2
File Description: application/pdf
Relation: https://www.malignanttumors.org/jour/article/view/1275/913; International Agency for Research on Cancer Global Cancer Observatory : Cancer Today. [(accessed on 5 June 2022)]; Available online: https://gco.iarc.fr/today/online-analysis-table [Ref list].; American Cancer Society Key Statistics for Ovarian Cancer. [(accessed on 5 June 2022)]. Available online: https://www.cancer.org/cancer/ovarian-cancer/about/key-statistics.html [Ref list].; Ghose A, Bolina A, Mahajan I, Raza SA, Clarke M, Pal A, et al. Hereditary Ovarian Cancer : Towards a Cost-Effective Prevention Strategy. Int J Environ Res Public Health. 2022 Sep 23; 19 (19) : 12057. doi:10.3390/ijerph191912057. PMID : 36231355; PMCID : PMC9565024.; Hinchcliff E.M., Bednar E., Lu K.H., Rauh-Hain J.A. Disparities in gynecologic cancer genetics evaluation. Gynecol. Oncol. 2019; 153 : 184–191. doi:10.1016/j.ygyno.2019.01.024.; Kanchi K.L., Johnson K.J., Lu C., McLellan M.D., Leiserson M.D.M., Wendl M.C., et al. Integrated analysis of germline and somatic variants in ovarian cancer. Nat. Commun. 2014; 5 : 3156. doi:10.1038/ncomms4156.; Biglia N., Sgandurra P., Bounous V.E., Maggiorotto F., Piva E., Pivetta E., et al. Ovarian cancer in BRCA1 and BRCA2 gene mutation carriers : Analysis of prognostic factors and survival. Ecancermedicalscience. 2016; 10 : 639. doi:10.3332/ecancer.2016.639.; Hunn J., Rodriguez G.C. Ovarian cancer : Etiology, risk factors, and epidemiology. Clin. Obstet. Gynecol. 2012; 55 : 3–23. doi:10.1097/GRF.0b013e31824b4611.; Andrews L, Mutch DG. Hereditary Ovarian Cancer and Risk Reduction. Best Pract Res Clin Obstet Gynaecol. 2017 May; 41 : 31–48. doi:10.1016/j.bpobgyn.2016.10.017. Epub 2017 Jan 17. PMID : 28254144.; Любченко Л. Н., Батенева Е. И. Медико-генетическое консультирование и ДНК-диагностика при наследственной предрасположенности к раку молочной железы и раку яичников .– м. : иг РОНЦ, 2014.; Alsop K, Fereday S, Meldrum C, deFazio A, Emmanuel C, George J, et al. BRCA mutation frequency and patterns of treatment response in BRCA mutation-positive women with ovarian cancer : a report from the Australian Ovarian Cancer Study Group. [Erratum appears in J Clin Oncol. 2012 Nov 20; 30 (33) : 4180]. Journal of Clinical Oncology 2012.30 (21) : 2654–63.; Bolton KL, Chenevix-Trench G, Goh C, Sadetzki S, Ramus SJ, Karlan BY, et al. Association between BRCA1 and BRCA2 mutations and survival in women with invasive epithelial ovarian cancer. JAMA 2012.307 (4) : 382–90.; Patch AM, Christie EL, Etemadmoghadam D, Garsed DW, George J, Fereday S, et al. Wholegenome characterization of chemoresistant ovarian cancer. Nature 2015.521 (7553) : 489–94.; https://www.malignanttumors.org/jour/article/view/1275
-
5Academic Journal
Authors: A. N. Letuchikh, A. S. Ablyametova, A. V. Zorinova, E. R. Israelyan, A. S. Tsareva, A. A. Rumyantsev, А. Н. Летучих, А. С. Абляметова, А. В. Зоринова, Э. Р. Исраелян, А. С. Царева, А. А. Румянцев
Contributors: The article was prepared without sponsorship., Статья подготовлена без спонсорской поддержки.
Source: Malignant tumours; Том 14, № 4 (2024); 84-90 ; Злокачественные опухоли; Том 14, № 4 (2024); 84-90 ; 2587-6813 ; 2224-5057
Subject Terms: определение аллелей партнера в опухолевом материале, non-gestational choriocarcinoma of the ovaries, gestational choriocarcinoma of the ovaries, molecular-genetic testing, determination of alleles of the partner in tumor material, негестационная хориокарцинома яичников, гестационная хориокарцинома яичников, молекулярно-генетическое тестирование
File Description: application/pdf
Relation: https://www.malignanttumors.org/jour/article/view/1324/1027; Bishop B.N., Edemekong P.F. Choriocarcinoma. In: StatPearls. Treasure Island (FL): StatPearls Publishing; February 4, 2023. PMID: 30571055; Liu X., Zhang X., Pang Y., et al. Clinicopathological factors and prognosis analysis of 39 cases of non-gestational ovarian choriocarcinoma. Arch Gynecol Obstet 2020;301(4):901–912. https://doi.org/10.1007/s00404-020-05502-9; Hayashi S., Abe Y., Tomita S., et al. Primary non-gestational pure choriocarcinoma arising in the ovary: A case report and literature review. Oncol Lett 2015;9(5):2109–2111. https://doi.org/10.3892/ol.2015.2985; Vance R.P., Geisinger K.R. Pure nongestational choriocarcinoma of the ovary. Report of a case. Cancer 1985;56(9):2321– 2325. https://doi.org/10.1002/1097-0142(19851101)56:93.0.co;2-q; Exman P., Takahashi T.K., Gattas G.F., et al. Primary ovary choriocarcinoma: individual DNA polymorphic analysis as a strategy to confirm diagnosis and treatment. Rare Tumors 2013;5(2):89–92. https://doi.org/10.4081/rt.2013.e24; Lv L., Yang K., Wu H., et al. Pure choriocarcinoma of the ovary: a case report. J Gynecol Oncol 2011;22(2):135–9. https://doi.org/10.3802/jgo.2011.22.2.135; Fisher R.A., Newlands E.S., Jeffreys A.J., et al. Gestational and nongestational trophoblastic tumors distinguished by DNA analysis. Cancer 1992;69(3):839–845. https://doi.org/10.1002/1097-0142(19920201)69:33.0.co;2-e; Adow M.T., Gebresilasie S.F., Abebe N.A. Primary ovarian choriocarcinoma: rare entity. Case Rep Obstet Gynecol 2021;2021:4545375. https://doi.org/10.1155/2021/4545375; Тюляндина А.С., Коломиец Л.А., Морхов К.Ю. и соавт. Практические рекомендации по лекарственному лечению рака яичников, первичного рака брюшины и рака маточных труб. Практические рекомендации RUSSCO, часть 1. Злокачественные опухоли 2023;13(3s2):201–215.; Гуторов С.Л., Новикова Е.Г., Румянцев А.А., и соавт. Практические рекомендации по лекарственному лечению злокачественных неэпителиальных опухолей яичников. Практические рекомендации RUSSCO, часть 1. Злокачественные опухоли 2023;13(3s2):216–234.; NCCN Guidelines, Version 1.2024. Gestational Trophoblastic Neoplasia. Available at: https://www.nccn.org/professionals/physician_gls/pdf/gtn.pdf; NCCN Guidelines, Version 1.2024. Testicular Cancer. Available at: https://www.nccn.org/professionals/physician_gls/pdf/testicular.pdf; Shao Y., Xiang Y., Jiang F., et al. Clinical features of a Chinese female nongestational choriocarcinoma cohort: a retrospective study of 37 patients. Orphanet J Rare Dis 2020;15(1):325. https://doi.org/10.1186/s13023-020-01610-6; Yu X., Du Q., Zhang X., et al. Pure primary non-gestational choriocarcinoma originating in the ovary: A case report and literature review. Rare Tumors 2021;13:20363613211052506. https://doi.org/10.1177/20363613211052506; Kong B., Tian Y.J., Zhu W.W., Qin Y.J. A pure nongestational ovarian choriocarcinoma in a 10-year-old girl: case report and literature review. J Obstet Gynaecol Res 2009;35(3):574–578. https://doi.org/10.1111/j.1447-0756.2008.00973.x; Lorigan P.C., Grierson A.J., Goepel J.R., et al. Gestational choriocarcinoma of the ovary diagnosed by analysis of tumour DNA. Cancer Lett 1996;104(1):27–30. https://doi.org/10.1016/0304-3835(96)04219-x; Zhang X., Yan K., Chen J., Xie X. Using short tandem repeat analysis for choriocarcinoma diagnosis: a case series. Diagn Pathol 2019;14:93. https://doi.org/10.1186/s13000-019-0866-5; Alifrangis C., Agarwal R., Short D., et al. EMA/CO for high-risk gestational trophoblastic neoplasia: good outcomes with induction low-dose etoposide-cisplatin and genetic analysis. J Clin Oncol 2013;31(2):280–286. https://doi.org/10.1200/JCO.2012.43.1817; You B., Bolze P.A., Lotz J.P., et al. Avelumab in patients with gestational trophoblastic tumors with resistance to single-agent chemotherapy: Cohort A of the TROPHIMMUN phase II trial. J Clin Oncol 2020;38(27):3129–3137. https://doi.org/10.1200/JCO.20.00803; You B., Bolze P.A., Lotz J.P., et al. Avelumab in patients with gestational trophoblastic tumors with resistance to polychemotherapy: Cohort B of the TROPHIMMUN phase 2 trial. Gynecol Oncol 2023;168:62–67. https://doi.org/10.1016/j.ygyno.2022.11.005; Yang Y., Zhang X., Chen D., et al. Adolescent non-gestational ovarian choriocarcinoma: report of a case and review of literature. Int J Clin Exp Pathol 2019;12(5):1788–1794. PMID: 31933999; Yamamoto E., Ino K., Yamamoto T., et al. A pure nongestational choriocarcinoma of the ovary diagnosed with short tandem repeat analysis: case report and review of the literature. Int J Gynecol Cancer 2007 Jan-Feb;17(1):254–258. https://doi.org/10.1111/j.1525-1438.2006.00764.x; Tsujioka H., Hamada H., Miyakawa T., et al. A pure nongestational choriocarcinoma of the ovary diagnosed with DNA polymorphism analysis. Gynecol Oncol 2003;89(3):540–542. https://doi.org/10.1016/s0090-8258(03)00139-2.; https://www.malignanttumors.org/jour/article/view/1324
-
6Academic Journal
Authors: V. А. Yarovaya, А. V. Golanov, V. V. Nazarova, А. R. Zaretskii, I. А. Levashov, А. К. Kulagina, Т. V. Melnikova, А. D. Matyaeva, А. А. Yarovoy, В. А. Яровая, А. В. Голанов, В. В. Назарова, А. Р. Зарецкий, И. А. Левашов, А. К. Кулагина, Т. В. Мельникова, А. Д. Матяева, А. А. Яровой
Source: Malignant tumours; Том 14, № 1 (2024); 83-91 ; Злокачественные опухоли; Том 14, № 1 (2024); 83-91 ; 2587-6813 ; 2224-5057
Subject Terms: клинический случай, fine needle aspiration biopsy, molecular testing, prognostic classes, Gamma Knife stereotactic radiosurgery, surveillance for metastatic disease, liver metastases, case report, тонкоигольная аспирационная биопсия, молекулярно-генетическое тестирование, прогностические классы, стереотаксическая радиохирургия «Гамма-нож», мониторинг метастатической болезни, метастазы в печени
File Description: application/pdf
Relation: https://www.malignanttumors.org/jour/article/view/1289/919; Яровой А.А., Голанов А.В., Ильялов С.Р. и соавт. Стереотаксическая радиохирургия «Гамма-нож» как альтернатива энуклеации у пациентов с увеальной меланомой (первые результаты). Офтальмохирургия 2014;2:74–80.; Яровой А.А., Демидов Л.В., Зарецкий А.Р., Яровая В.А., Назарова В.В., Чудакова Л.В. и др. Способ определения тактики ведения пациентов с увеальной меланомой: Патент № 2019136524. 2019; Singh A.D., Bergman L., Seregard S. Uveal melanoma: epidemiologic aspects. Ophthalmol Clin North Am 2005;18(1):75– 84. https://doi.org/10.1016/j.ohc.2004.07.002; Damato E.M., Damato B.E. Detection and time to treatment of uveal melanoma in the United Kingdom: an evaluation of 2384 patients. Ophthalmology 2012;119(8):1582–9. https://doi.org/10.1016/j.ophtha.2012.01.048; Яровой А.А., Малюгин Э.Б., Яровая В.А. и соавт. Тонкоигольная аспирационная биопсия внутриглазных образований. Офтальмохирургия 2020;1:51–56. https://doi.org/10.25276/0235-4160-2020-1-51-56.; Margo C.E. The collaborative ocular melanoma study: an overview. Cancer Control 2004;11(5):304–309. https://doi.org/10.1177/107327480401100504; Яровой А.А. Органосохраняющее и функционально-сберегающее лечение меланомы хориоидеи на основе брахитерапии с рутением-106 и лазерной транспупиллярной термотерапии. Дис. . д-ра мед. наук. М., 2010.; Shah A.A., Bourne T.D., Murali R. BAP1 protein loss by immunohistochemistry: a potentially useful tool for prognostic prediction in patients with uveal melanoma. Pathology 2013;45(7):651–6. https://doi.org/10.1097/PAT.0000000000000002; Damato B., Kacperek A., Errington D., Heimann H. Proton beam radiotherapy of uveal melanoma. Saudi J Ophthalmol 2013;27(3):151–157. https://doi.org/10.1016/j.sjopt.2013.06.014; Саакян С.В., Бородин Ю.И., Ширина Т.В. Оценка эффективности лечения и выживаемости больных увеальной меланомой после лечения медицинским узким протонным пучком. Радиология-практика 2012;6:49–53; Синявский О.А., Трояновский Р.Л., Иванов П.И. и соавт. Пятилетний опыт резекции увеальных меланом после стереотаксической радиохирургии с использованием гамма-ножа. Современные технологии в офтальмологии 2018;1:316–319.; Parker T., Rigney G., Kallos J., et al. Gamma knife radiosurgery for uveal melanomas and metastases: a systematic review and meta-analysis. Lancet Oncol 2020;21(11):1526–1536. https://doi.org/10.1016/S1470-2045(20)30459-9; Shields C.L., Say E.A. T., Hasanreisoglu M., et al. Personalized prognosis of uveal melanoma based on cytogenetic profile in 1059 patients over an 8-year period: the 2017 Harry S. Gradle Lecture. Ophthalmology 2017;124(10):1523– 1531. https://doi.org/10.1016/j.ophtha.2017.04.003; Cicinelli M.V., Nicola M.D., Gigliotti C.R., et al. Predictive factors of radio-induced complications in 194 eyes undergoing gamma knife radiosurgery for uveal melanoma. Acta Ophthalmol 2021;99(8):e1458-e1466. https://doi.org/10.1111/aos.14814; Яровая В.А. Прогностическая тонкоигольная аспирационная биопсия увеальной меланомы. Дис. . канд. ме. наук. М., 2020.; Kilic E., Naus N.C., van Gils W., et al. Concurrent loss of chromosome arm 1p and chromosome 3 predicts a decreased disease-free survival in uveal melanoma patients. Invest Ophthalmol Vis Sci 2005;46(7):2253–7. https://doi.org/10.1167/iovs.04-1460; Singh N., Bergman L., Seregard S., Singh A.D. Uveal melanoma: epidemiologic aspects. Clinical Ophthalmic Oncology. Uveal Tumors 2014:75–88. https://doi.org/10.1007/978-3-642-54255-8_6; Carvajal R.D., Schwartz G.K., Tezel T., et al. Metastatic disease from uveal melanoma: treatment options and future prospects. Br J Ophthalmol 2017;101(1):38–44. https://doi.org/10.1136/bjophthalmol-2016-309034; https://www.malignanttumors.org/jour/article/view/1289
-
7Academic Journal
Authors: O. P. Nechay, O. A. Tovkai, V. O. Palamarchuk, N. I. Belemets, S. І. Nikolayenko, O. V. Mazur, D. M. Kvitka, P. O. Lishchynsky
Source: Clinical Endocrinology and Endocrine Surgery; No. 2 (2021); 9-16
Clinical Endocrinology and Endocrine Surgery; № 2 (2021); 9-16Subject Terms: thyroid cancer, molecular genetic testing, gene, рак щитоподібної залози, молекулярно-генетичне тестування, ген, рак щитовидной железы, молекулярно-генетическое тестирование, 3. Good health
File Description: application/pdf
-
8
-
9Academic Journal
Authors: T. E. Tikhomirova, A. S. Tyulyandina, A. A. Rumyantsev, M. E. Abramov, A. Yu. Anokhin, A. N. Lud, S. A. Tjulandin, Т. Е. Тихомирова, А. С. Тюляндина, А. А. Румянцев, М. Е. Абрамов, А. Ю. Анохин, А. Н. Луд, С. А. Тюляндин
Source: Surgery and Oncology; Том 12, № 3 (2022); 56-62 ; Хирургия и онкология; Том 12, № 3 (2022); 56-62 ; 2949-5857
Subject Terms: молекулярно-генетическое тестирование, mutations in BRCA1 / 2 genes, molecular genetic testing, мутации в генах BRCA1 / 2
File Description: application/pdf
Relation: https://www.onco-surgery.info/jour/article/view/554/399; Webb P.M., Jordan S.J. Epidemiology of epithelial ovarian cancer. Best Pract Res Clin Obstet Gynaecol 2017;41:3–14. DOI:10.1016/j.bpobgyn.2016.08.006; American Cancer Society – Cancer Facts and Figures 2019. Available at: https://www.cancer.org/research/cancer-factsstatistics/all-cancer-facts-figures/cancer-facts-figures-2019.html.; American Cancer Society. Cancer Statistics Center. Available at: http://cancerstatisticscenter.cancer.org.; McCluggage W.G. Morphological subtypes of ovarian carcinoma: a review with emphasis on new developments and pathogenesis. Pathology 2011;43(5):420–32. DOI:10.1097/PAT.0b013e328348a6e7; Невмержицкая Н.Н., Дзевульская И.В., Безштанько Н.А. и др. Выдающийся Поль Брокка (к 140-летию со дня памяти). Вестник проблем биологии и медицины 2020;1(155):12–6. DOI:10.29254/2077-4214-2020-1-155-12-16; Hall J.M., Lee M.K., Newman B. et sl. Linkage of early-onset familial breast cancer to chromosome 17q21. Science 1990;250(4988):1684–9. DOI:10.1126/science.2270482; Miki Y., Swensen J., Shattuck-Eidens D. et al. a strong candidate for the breast and ovarian cancer susceptibility gene BRCA1. Science 1994;266(5182):66–71. DOI:10.1126/science.7545954; Wooster R., Neuhausen S.L., Mangion J. et al. Localization of a breast cancer susceptibility gene, BRCA2, to chromosome 13q12-13. Science 1994;265(5181):2088–90. DOI:10.1126/science.8091231; Lord C.J., Ashworth A. the DNA damage response and cancer therapy. Nature 2012;481:287–94. DOI:10.1038/nature10760; Kuchenbaecker K.B., Hopper J.L., Barnes D.R. et al. Risks of breast, ovarian, and contralateral breast cancer for BRCA1 and BRCA2 mutation carriers. JAMA 2017;317(23):2402–16. DOI:10.1001/jama.2017.7112; Yoshida R. Hereditary breast and ovarian cancer (HBOC): review of its molecular characteristics, screening, treatment, and prognosis. Breast Cancer 2021;28(6):1167–80. DOI:10.1007/s12282-020-01148-2; Antoniou A., Pharoah P.D., Narod S. et al. Average risks of breast and ovarian cancer associated with BRCA1 or BRCA2 mutations detected in case series unselected for family history: a combined analysis of 22 studies. Am J Hum Genet 2003;72:1117–30. DOI:10.1086/375033; Chen S., Parmigiani G. Meta-analysis of BRCA1 and BRCA2 penetrance. J Clin Oncol 2007;10(25):1329–33. DOI:10.1200/JCO.2006.09.1066; Kang E., Kim S.W. the Korean hereditary breast cancer study: review and future perspective. J Breast Cancer 2013;16:245–53. DOI:10.4048/jbc.2013.16.3.245; Eccles D.M., Balmaña J., Clune J. et al. Selecting patients with ovarian cancer for germline BRCA mutation testing: findings from guidelines and a systematic literature review. Adv Ther 2016;33(2):129–50. DOI:10.1007/s12325-016-0281-1; Armstrong D.K., Alvarez R.D., Bakkum-Gamez J.N. et al. Ovarian Cancer, Version 2.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2021;19(2): 191–226. DOI:10.6004/jnccn.2021.0007; Любченко Л.Н., Батенева Е.И. Медико-генетическое консультирование и ДНК-диагностика при наследственной предрасположенности к раку молочной железы и раку яичников. Пособие для врачей. М.: ИГ РОНЦ, 2014. 64 с.; Finch A.P., Lubinski J., Møller P. et al. Impact of oophorectomy on cancer incidence and mortality in women with a BRCA1 or BRCA2 mutation. J Clin Oncol 2014;32(15):1547–53. DOI:10.1200/JCO.2013.53.2820; Alsop K., Fereday S., Meldrum C. et al. BRCA mutation frequency and patterns of treatment response in BRCA mutation-positive women with ovarian cancer: a report from the Australian Ovarian Cancer Study Group. J Clin Oncol 2012;30(21):2654–63. DOI:10.1200/JCO.2011.39.8545. Erratum in: J Clin Oncol 2012;30(33):4180.; Struewing J.P., Hartge P., Wacholder S. et al. The risk of cancer associated with specific mutations of BRCA1 and BRCA2 among Ashkenazi Jews. N Engl J Med 1997;336(20):1401–8. DOI:10.1056/NEJM199705153362001; Соколенко А.П., Розанов М.Е., Митюшкина Н.В. и др. Наследственные мутации при ранних, семейных и билатеральных формах рака молочной железы у пациенток из России. Сибирский онкологический журнал 2008;(3):43–9.; Safra T., Lai W.C., Borgato L. et al. BRCA mutations and outcome in epithelial ovarian cancer (EOC): experience in ethnically diverse groups. Ann Oncol 2013;24(Suppl 8):viii63–viii68. DOI:10.1093/annonc/mdt315; Бровкина О.И., Гордиев М.Г., Еникеев Р.Ф. и др. Гены системы репарации: популяционные различия наследственных типов рака яичников и молочной железы, выявляемые методом секвенирования нового поколения. Опухоли женской репродуктивной системы 2017;13(2):61–7. DOI:10.17650/1994-4098-2017-13-2-61-67; Janavičius R. Founder BRCA1/2 mutations in the Europe: implications for hereditary breast-ovarian cancer prevention and control. EPMA J 2010;1(3):397–412. DOI:10.1007/s13167-010-0037-y; Singer C.F., Tan Y.Y., Muhr D. et al. Association between family history, mutation locations, and prevalence of BRCA1 or 2 mutations in ovarian cancer patients. Cancer Med 2019;8(4):1875–81. DOI:10.1002/cam4.2000; Саевец В.В., Тюляндина А.С., Кекеева Т.В. и др. Финальный анализ неинтервенционного исследования OVATAR: диагностические и лечебные подходы к лечению рака яичников в России. Анализ группы с мутациями BRCA. Злокачественные опухоли 2019;9(3S1):90, 91.; Pennington K.P., Walsh T., Harrell M.I. et al. Germline and somatic mutations in homologous recombination genes predict platinum response and survival in ovarian, fallopian tube, and peritoneal carcinomas. Clin Cancer Res 2014;20(3):764–75. DOI:10.1158/1078-0432.CCR-13-2287; Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 2011;474(7353):609–15. DOI:10.1038/nature10166. Erratum in: Nature 2012;490(7419):298.; Hahnen E., Baumann K., Heimbach A. et al. Prevalence of somatic mutations in risk genes including BRCA1/2 in consecutive ovarian cancer patients (AGO-TR-1 study). J Clin Oncol 2016;34:5544.; Boyd J., Sonoda Y., Federici M.G. et al. Clinicopathologic features of BRCA-linked and sporadic ovarian cancer. JAMA 2000;283(17):2260–5. DOI:10.1001/jama.283.17.2260; Pan Z., Xie X. BRCA mutations in the manifestation and treatment of ovarian cancer. Oncotarget 2017;8(57):97657–70. DOI:10.18632/oncotarget.18280; Mavaddat N., Barrowdale D., Andrulis I.L. et al. Consortium of Investigators of Modifiers of BRCA1/2. Pathology of breast and ovarian cancers among BRCA1 and BRCA2 mutation carriers: results from the Consortium of Investigators of Modifiers of BRCA1/2 (CIMBA). Cancer Epidemiol Biomarkers Prev 2012;21(1):134–47. DOI:10.1158/1055-9965.EPI-11-0775; Aoki D., Chiyoda T. PARP inhibitors and quality of life in ovarian cancer. Lancet Oncol 2018;19(8):1012–4. DOI:10.1016/S1470-2045(18)30435-2; Moore K., Colombo N., Scambia G. et al. Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med 2018;379(26):2495–505. DOI:10.1056/NEJMoa1810858; Pujade-Lauraine E., Ledermann J.A., Selle F. et al. SOLO2/ ENGOT-Ov21 investigators. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol 2017;18(9):1274–84. DOI:10.1016/S1470-2045(17)30469-2
-
10Academic Journal
-
11Academic Journal
Source: Clinical Endocrinology and Endocrine Surgery; No. 2 (2021); 9-16
Clinical Endocrinology and Endocrine Surgery; № 2 (2021); 9-16Subject Terms: thyroid cancer, molecular genetic testing, gene, рак щитоподібної залози, молекулярно-генетичне тестування, ген, рак щитовидной железы, молекулярно-генетическое тестирование, 3. Good health
File Description: application/pdf
-
12Academic Journal
Authors: РОМАНОВА В.В.
File Description: text/html
-
13Academic Journal
Source: Приволжский научный вестник.
Subject Terms: 2. Zero hunger, СИММЕНТАЛЫ МЕСТНОЙ СЕЛЕКЦИИ,ПРОДУКТИВНОЕ ДОЛГОЛЕТИЕ,ОЦЕНКА БЫКОВ ПО КАЧЕСТВУ ПОТОМСТВА,МОЛЕКУЛЯРНО-ГЕНЕТИЧЕСКОЕ ТЕСТИРОВАНИЕ,SIMMENTAL COWS OF LOCAL SELECTION,PRODUCTIVE LONGEVITY,ESTIMATION OF BULLS ON QUALITY OF POSTERITY,MOLECULAR-GENETIC TESTING
File Description: text/html
-
14Report
Subject Terms: ген SPG11, hypoplasia of corpus callosum, наследственная спастическая параплегия 11-го типа, спатасцин, hereditary spastic paraplegia type 11, spatascin, 3. Good health, гемипаркинсонизм, SPG11 gene, hemiparkin- sonism, lower paraparesis, molecular ge- netic testing, истончение мозолистого тела, молекулярно-генетическое тестирование, нижний парапарез