-
1Academic Journal
المؤلفون: Uralov Bakhtiyor, Eshev Sobir, Khakimova G., Mutalov Sh., Raimova I., Arzieva D., Salimbayev Mirsoxibjon
المصدر: E3S Web of Conferences, Vol 401, p 01052 (2023)
مصطلحات موضوعية: Environmental sciences, GE1-350
وصف الملف: electronic resource
-
2Academic Journal
المؤلفون: Ovsyannikova, I V, Asfandiyarova, L R, Khakimova, G V, Luzina, M S
المصدر: IOP Conference Series: Earth and Environmental Science ; volume 1231, issue 1, page 012050 ; ISSN 1755-1307 1755-1315
-
3Conference
المؤلفون: Shulaev, N. S., Pryanichnikova, V. V., Ovsyannikova, I. V., Kadyrov, R. R., Khakimova, G. V., Asfandiyarova, L. R.
المصدر: AIP Conference Proceedings ; PROCEEDINGS OF THE 2ND INTERNATIONAL INTERDISCIPLINARY SCIENTIFIC CONFERENCE “DIGITALIZATION AND SUSTAINABILITY FOR DEVELOPMENT MANAGEMENT: ECONOMIC, SOCIAL, AND ENVIRONMENTAL ASPECTS” ; volume 3033, page 030017 ; ISSN 0094-243X
-
4Conference
المؤلفون: Ovsyannikova, I. V., Asfandiyarova, L. R., Khakimova, G. V.
المصدر: AIP Conference Proceedings ; PROCEEDINGS OF THE 2ND INTERNATIONAL INTERDISCIPLINARY SCIENTIFIC CONFERENCE “DIGITALIZATION AND SUSTAINABILITY FOR DEVELOPMENT MANAGEMENT: ECONOMIC, SOCIAL, AND ENVIRONMENTAL ASPECTS” ; volume 3033, page 030015 ; ISSN 0094-243X
-
5Academic Journal
المؤلفون: Khоlikova S., Ruziev Sh., Khakimova G.
المصدر: Znanstvena misel journal, 66, 43-49, (2022-05-23)
مصطلحات موضوعية: hydroquinone, urea, thiourea, cyclocondensation,indole, synthesis, ethylene glycol, one-step method, aniline, catalysts, heterocyclization, N-aminophenylethanol, N-vinylaniline
Relation: https://doi.org/10.5281/zenodo.6572650; https://doi.org/10.5281/zenodo.6572651; oai:zenodo.org:6572651
-
6Academic Journal
المؤلفون: Asfandiyarova, L R, Khakimova, G V, Il’in, V M
المصدر: IOP Conference Series: Earth and Environmental Science ; volume 1076, issue 1, page 012009 ; ISSN 1755-1307 1755-1315
-
7Academic Journal
المؤلفون: Khakimova, G V, Asfandiyarova, L R
المصدر: IOP Conference Series: Earth and Environmental Science ; volume 981, issue 3, page 032074 ; ISSN 1755-1307 1755-1315
-
8Academic Journal
المؤلفون: A. Zikiryakhodzhaev D., Sh. Khakimova G., E. Rasskazova A., E. Saribekyan K., D. Omarova F., V. Surkova S., G. Khakimova G., А. Зикиряходжаев Д., Ш. Хакимова Г., Е. Рассказова А., Э. Сарибекян К., Д. Омарова Ф., В. Суркова С., Г. Хакимова Г.
المصدر: Malignant tumours; Том 10, № 4 (2020); 30-37 ; Злокачественные опухоли; Том 10, № 4 (2020); 30-37 ; 2587-6813 ; 2224-5057
مصطلحات موضوعية: breast cancer, squamous cell breast cancer, lymph node dissection, histopathological examination, mesh implant, combination therapy, рак молочной железы, плоскоклеточныйрак молочнойжелезы, лимфаденэктомия, гистологическое исследование, сетчатый имплантант, комбинированное лечение
وصف الملف: application/pdf
Relation: https://www.malignanttumors.org/jour/article/view/826/580; Paletta C, Paletta FX, Jr., Paletta FX, Sr. Squamous cell carcinoma following breast augmentation. Ann Plast Surg 1992; 29, 425–429; discussion 429–432; Molitor M, Mestak O, Kalinova L, Krajcova A, Mestak J. The history and safety of breast implants. Acta Chir Plast. 2014;56 (1-2):15–19.; Patrick J. Buchanan, Vinod K. Chopra, Kristen L. Walker Primary Squamous Cell Carcinoma Arising from a Breast Implant Capsule: A Case Report and Review of the Literature The American Society for Aesthetic Plastic Surgery –2018; SmithLF, SmithTT, YearyE, McGeeJM, MalnarK: Squamouscellcarcinomaofthebreast following silicone injection of the breasts. J Okla State Med Assoc. 1999, 92:126–130.; Barr LH, Menard JW: Marjolin’s ulcer. The LSU experience. Cancer. 1983, 52:173–175.10.1002/1097–0142 (19830701) 52:13.0. CO;2–6; Paletta C, Paletta FX Jr, Paletta FX Sr: Squamous cell carcinoma following breast augmentation. Ann Plast Surg. 1992, 29:425–429.; Satgunaseelan L, Cheung D, Reddy J: Breast implant-associated squamous cell carcinoma—a rare long term complication. Pathology. 2015, 47:72–73. 10.1097/01. PAT. 0000461520.64127.87; Zomerlei TA, Samarghandi A, Terando AM: Primary squamous cell carcinoma arising from a breast implant capsule. Plast Reconstr Surg Glob Open. 2015, 3:586. 10.1097/GOX. 0000000000000567; Olsen DL, Keeney GL, Chen B, Visscher DW, Carter JM: Breast implant capsule-associated squamous cell carcinoma: a report of 2 cases. Hum Pathol. 2017, 67:94–100. 10.1016/j. humpath. 2017.07.011; Zhou YM, Chaudhry HE, Shah A, Andrews J. Breast Squamous Cell Carcinoma Following Breast Augmentation. Cureus. 2018;10 (10):e3405. Published 2018 Oct 3. doi:10.7759/cureus. 3405; Handel N, Cordray T, Gutierrez J, Jensen JA: A long-term study of outcomes, complications, and patient satisfaction with breast implants. Plast Reconstr Surg. 2006, 117:757–767. 10.1097/01. prs. 0000201457.00772.1d; Rosen PP, Hoda SA, Brogi E, Koerner FC: Rosen’s Breast Pathology. Wolters Kluwer, Philadelphia; 2014.; Yadav S, Yadav D, Zakalik D: Squamous cell carcinoma of the breast in the United States: incidence, demographics, tumor characteristics, and survival. Breast Cancer Res Treat. 2017, 164:201–208. 10.1007/s10549–017–4251–3; Lakhani SR: WHO classification of tumours of the breast: [views of a working group that convened for a consensus and editorial meeting at the International Agency for Research on Cancer]. International Agency for Research on Cancer. World Health Organization, Lyon; 2012. 1:430.; Anne N. Primary squamous cell carcinoma of the breast: a case report and review of the literature/N. Anne, E. Sulger, R. Pallapothu// J Surg Case Rep. –2019.—Vol. 6.—P. 182. doi:10.1093/jscr/rjz182; https://www.malignanttumors.org/jour/article/view/826
-
9Academic Journal
المؤلفون: G. Khakimova G., A. Tryakin A., T. Zabotina N., Sh. Khakimova G., Г. Хакимова Г., А. Трякин А., Т. Заботина Н., Ш. Хакимова Г.
المصدر: Malignant tumours; Том 10, № 4 (2020); 5-15 ; Злокачественные опухоли; Том 10, № 4 (2020); 5-15 ; 2587-6813 ; 2224-5057
مصطلحات موضوعية: gastric adenocarcinoma, cellular immunity, local immunity, subpopulation of lymphocytes, neutrophil‑ lymphocyte index, platelet‑lymphocyte index, аденокарцинома желудка, клеточный иммунитет, локальный иммунитет, субпопуляция лимфоцитов, нейтрофильно‑лимфоцитраный индекс, тромбоцитарно‑лимфоцитарный индекс
وصف الملف: application/pdf
Relation: https://www.malignanttumors.org/jour/article/view/823/577; Бережная Н. М. Взаимодействие клеток системы иммунитета с другими компонентами микроокружения. Онкология. 2009; 1 (2): 86–93.; Mantovani A. et al. Tumor immunity: effector response to tumor and role of the microenvironment. Lancet 2008; 371 (9614): 771–83.; Тупицын Н. Н. Иммунофенотип рака молочнойжелезы. В кн.: Рак молочнойжелезы. Под ред. Н. Е. Кушлинского, С.М. Портного, К. П. Лактионова. М.: Издательство РАМН, 2005; с. 174–97.; Balch C., Riley L., Bae T. et al. Patterns of human tumor infiltrating lymphocytes in 120 human cancers. Arch Surg 1990; 125 (2): 200–5.; Galon J., Pages F. et al. Cancer classification using the immunoscore: a worldwide task force. J Transl Med 2012; 10: 205.; Zamarron B. F., Chen W. Dual roles of immune cells and their factors in cancer development and progression. Int J Biol Sci. 2011; 7 (5): 651–658.; Ruffell B., DeNardo D. G., Affara N. I. et al. Lymphocytes in cancer development: polarization towards protumor immunity. Cytokine Growth Factor Rev. 2010; 21: 3–10.; Shen Z. Zhou S., Wang Y. et al. Higher intratumoral infiltrated Foxp3 + Treg numbers and Foxp3 + / C8 + ratio are associated with adverse prognosis in resectable gastric cancer. J Cancer Res Clin. Oncol. 2010; 136:1585–1595.; Mori M., Shuto K., Kosugi C. et al. An increase in the neutrophil to lymphocyte ratio during adjuvant chemotherapy indicates a poor prognosis in patients with stage II or III gastric cancer. BMC Cancer. 2018;18 (1):1261.; Passardi A., Scarpi E., Cavanna L. et al. Inflammatory indexes as predictors of prognosis and bevacizumab efficacy in patients with metastatic colorectal cancer. Oncotarget 2016;7 (22):33210–9. DOI:10.18632 / oncotarget. 8901.; Gunaldi M., Goksu S., Erdem D. et al. Prognostic impact of platelet / lymphocyte and neutrophil / lymphocyte ratios in patients with gastric cancer: a multicenter study. Int J Clin Exp Med. 2015;8 (4):5937–42.; Zheng X., Song X., Shao Y. et al. Prognostic role of tumor-infiltrating lymphocytes in gastric cancer: A meta-analysis. Oncotarget. 2017; 8. 10.18632 / oncotarget. 18065.; Cancer Genome Atlas Research N. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014; 513: 202–209.; Müller P., Rothschild S. I., Arnold W. et al. Metastatic spread in patients with non-small cell lung cancer is associated with a reduced density of tumor-infiltrating T cells. Cancer Immunol Immunother 2016; 65: 1–1.; Kollmann D., Ignatova D., Jedamzik J. et al. Expression of programmed cell death protein 1 by tumor-infiltrating lymphocytes and tumor cells is associated with advanced tumor stage in patients with esophageal adenocarcinoma. Ann Surg Oncol 2017; 24: 2698–706.; Huszno J., Nożyńska E. Z., Lange D. et al. The association of tumor lymphocyte infiltration with clinicopathological factors and survival in breast cancer. Pol J Pathol 2017; 68: 26–32.; Lu J., Xu Y., Wu Y. et al. Tumor-infiltrating CD8 + T cells combined with tumor-associated CD68 + macrophages predict postoperative prognosis and adjuvant chemotherapy benefit in resected gastric cancer. BMC Cancer 2019; 920 (19).; Ebihara T., Sakai N., Koyama S. Suppression by sorted CD8 + CD11b-cells from T-cell growth factor-activated peripheral blood lymphocytes on cytolytic activity against tumor in patients with gastric carcinoma. Eur J of Cancer 1991; 27 (12): 1654–7 / 19. Hou J., Yu Z., Xiang R. et al. Correlation between infiltration of FOXP3 + regulatory T cells and expression of B7-H1 in the tumor tissues of gastric cancer. Exp Molecular Pathol 2014; 96 (3): 284–91.; Yuan X. L., Chen L., Li M-X. et al. Elevated expression of Foxp3 in tumor-infiltrating Treg cells suppresses T-cell proliferation and contributes to gastric cancer progression in a COX - 2-dependent manner. Clin Immunol 2010; 134 (3): 277–88.; Peng L. S., Mao F. Y., Zhao Y. L. et al. Altered phenotypic and functional characteristics of CD3 + CD56 + NKT-like cells in human gastric cancer. Oncotarget 2016; 7 (34): 55222–30. DOI:10.18632 / oncotarget. 10484; Takeuchi H., Maehara Y., Tokunaga E. et al. Prognostic Significance of Natural Killer Cell Activity in Patients With Gastric Carcinoma: A Multivariate Analysis. Am J Gastroenterol 2001; 96 (2): 574–8.; https://www.malignanttumors.org/jour/article/view/823
-
10Academic Journal
المؤلفون: Asfandiyarova, L R, Khakimova, G V, Ovsyannikova, I V
المصدر: IOP Conference Series: Earth & Environmental Science; 2024, Vol. 1390 Issue 1, p1-5, 5p
-
11Academic Journal
المؤلفون: I. Duadze S., A. Zikiryakhodzhaev D., A. Sukhotko S., M. Starkova V., F. Usov N., D. Bagdasarova V., D. Dzhabrailova Sh., Sh. Khakimova G., И. Дуадзе С., А. Зикиряходжаев Д., А. Сухотько С., М. Старкова В., Ф. Усов Н., Д. Багдасарова В., Д. Джабраилова Ш., Ш. Хакимова Г.
المصدر: Research and Practical Medicine Journal; Том 8, № 3 (2021); 108-117 ; Research'n Practical Medicine Journal; Том 8, № 3 (2021); 108-117 ; 2410-1893 ; 2409-2231 ; 10.17709/2410-1893-2021-8-3
مصطلحات موضوعية: breast cancer, breast reconstruction, DIEP-flap, TRAM-flap, perforant flaps, autologous flap, рак молочной железы, реконструкция молочной железы, DIEP-лоскут, TRAM-лоскут, перфорантные лоскуты, аутологичные лоскуты
وصف الملف: application/pdf
Relation: https://www.rpmj.ru/rpmj/article/view/702/450; https://www.rpmj.ru/rpmj/article/downloadSuppFile/702/560; Каприн А.Д., Старинский В.В., Петрова Г.В. Злокачественные новообразования в России в 2018 году (заболеваемость и смертность). М.: ФГБУ «МНИОИ им. П.А. Герцена» Минздрава России, 2019, 44 с. Доступно по: https://glavonco.ru/cancer_register/Забол_2018_Электр.pdf. Дата обращения: 10.07.2021; Merino Bonilla JA, Torres Tabanera M, Ros Mendoza LH. Breast cancer in the 21st century: from early detection to new therapies. Radiologia. 2017 Oct;59(5):368–379. https://doi.org/10.1016/j.rx.2017.06.003; Суздальцев И.В., Блохин С.Н., Надеин К.В. Реконструктивно-пластические операции после радикального лечения рака молочных желез Медицинский вестник Северного Кавказа. 2012;(3):105–110. Доступно по: https://www.elibrary.ru/download/elibrary_17992917_17772500.pdf. Дата обращения: 22.07.2021; Боровиков А.М. Восстановление груди после мастэктомии. М.: «Губернская медицина» 2000, 287 с. Доступно по: https://rusneb.ru/catalog/004796_000040_TVERS-RU%7C%7C%7CTOUNB%7C%7C%7CEKOLD%7C%7C%7C5-8376-0014-0/ Дата обращения: 05.07.2021; Tansini I. Sopra il mio processo di amputazione della mammella. Gazz Med Ital 1906;57:141. Доступно на: https://www.scienceopen.com/document?vid=f7f92290-2c3b-451e-b501-dca7a59f083e Дата обращения: 12.07.2021; Olivari N. The latissimus flap. Br J Plast Surg. 1976 Apr;29(2):126–128. https://doi.org/10.1016/0007-1226(76)90036-9; Holmstrom H. The free abdominoplasty flap and its use in breast reconstruction. An experimental study and clinical case report. Scand J Plast Reconstr Surg. 1979;13(3):423–427. https://doi.org/10.3109/02844317909013092; Hartrampf CR, Scheflan M, Black PW. Breast reconstruction with a transverse abdominal island flap. Plast Reconstr Surg. 1982 Feb;69(2):216–225. https://doi.org/10.1097/00006534-198202000-00006; Эйзенманн-Клайн М., Нейханн-Лоренц К. Пластическая и эстетическая хирургия: последние достижения. Перевод с английского под редакцией А. М. Боровикова. М.: Практическая медицина, 2011, 446 с.; Buck DW, Fine NA. The pedicled transverse rectus abdominis myocutaneous flap: indications, techniques, and outcomes. Plast Reconstr Surg. 2009 Oct;124(4):1047–1054. https://doi.org/10.1097/PRS.0b013e3181b457b2; Nahabedian M, Neligan P. Plastic surgery 4th edition. Elsevier; 2017, 576 p. Доступно по: https://www.elsevier.com/books/plastic-surgery/nahabedian/978-0-323-35709-8 Дата обращения: 12.07.2021; Allen RJ, Treece P. Deep inferior epigastric perforator flap for breast reconstruction. Ann Plast Surg. 1994 Jan;32(1):32–38. https://doi.org/10.1097/00000637-199401000-00007; Blondeel PN. One hundred free DIEP flap breast reconstructions: a personal experience. Br J Plast Surg. 1999 Mar;52(2):104–111. https://doi.org/10.1054/bjps.1998.3033; Tan MG, Isaranuwatchai W, DeLyzer T, Butler K, Hofer SOP, O’Neill AC, et al. A cost-effectiveness analysis of DIEP vs free MS-TRAM flap for microsurgical breast reconstruction. J Surg Oncol. 2019 Mar;119(3):388–396. https://doi.org/10.1002/jso.25325; Ludolph I, Horch RE, Harlander M, Arkudas A, Bach AD, Kneser U, et al. Is there a Rationale for Autologous Breast Reconstruction in Older Patients? A Retrospective Single Center Analysis of Quality of life, Complications and Comorbidities after DIEP or ms-TRAM Flap Using the BREAST-Q. Breast J. 2015 Dec;21(6):588–595. https://doi.org/10.1111/tbj.12493; American Cancer Society, Surveillance Research. Cancer Facts & Figures, 2019, 76 с. Доступно по: https://www.cancertutor.com/wp-content/uploads/2019/03/cancer-facts-and-figures-2019.pdf. Дата обращения: 15.06.2021; Ng SK, Hare RM, Kuang RJ, Smith KM, Brown BJ, Hunter-Smith DJ. Breast Reconstruction Post Mastectomy: Patient Satisfaction and Decision Making. Ann Plast Surg. 2016 Jun;76(6):640–644. https://doi.org/10.1097/SAP.0000000000000242; Boughey JC, Attai DJ, Chen SL, Cody HS, Dietz JR, Feldman SM, et al. Contralateral Prophylactic Mastectomy (CPM) Consensus Statement from the American Society of Breast Surgeons: Data on CPM Outcomes and Risks. Ann Surg Oncol. 2016 Oct;23(10):3100–3105. https://doi.org/10.1245/s10434-016-5443-5; Leyngold MM. Is Unipedicled Transverse Rectus Abdominis Myocutaneous Flap Obsolete Owing to Superiority of DIEP Flap? Ann Plast Surg. 2018 Jun;80(6S Suppl 6):S418–S420. https://doi.org/10.1097/SAP.0000000000001319; Seidenstuecker K, van Waes C, Munder BI, Claes KEY, Witzel C, Roche N, et al. DIEAP flap for safe definitive autologous breast reconstruction. Breast. 2016 Apr;26:59–66. https://doi.org/10.1016/j.breast.2015.12.005; Saint-Cyr M, Chang DW, Robb GL, Chevray PM. Internal mammary perforator recipient vessels for breast reconstruction using free TRAM, DIEP, and SIEA flaps. Plast Reconstr Surg. 2007 Dec;120(7):1769–1773. https://doi.org/10.1097/01.prs.0000287132.35433.d6; Jeong W, Lee S, Kim J. Meta-analysis of flap perfusion and donor site complications for breast reconstruction using pedicled versus free TRAM and DIEP flaps. Breast. 2018 Apr;38:45–51. https://doi.org/10.1016/j.breast.2017.12.003; Grünherz L, Keijzer W, Uyulmaz S, Fertsch S, Imhof L, Käser S, et al. Donor site aesthetics and morbidity after DIEP flap breast reconstruction-A retrospective multicenter study. Breast J. 2020 Oct;26(10):1980–1986. https://doi.org/10.1111/tbj.14003; Alderman AK, Atisha D, Streu R, Salem B, Gay A, Abrahamse P, et al. Patterns and correlates of postmastectomy breast reconstruction by U.S. Plastic surgeons: results from a national survey. Plast Reconstr Surg. 2011 May;127(5):1796–1803. https://doi.org/10.1097/PRS.0b013e31820cf183; Hu ES, Pusic AL, Waljee JF, Kuhn L, Hawley ST, Wilkins E, et al. Patient-reported aesthetic satisfaction with breast reconstruction during the long-term survivorship Period. Plast Reconstr Surg. 2009 Jul;124(1):1–8. https://doi.org/10.1097/PRS.0b013e3181ab10b2; Macadam SA, Zhong T, Weichman K, Papsdorf M, Lennox PA, Hazen A, et al. Quality of Life and Patient-Reported Outcomes in Breast Cancer Survivors: A Multicenter Comparison of Four Abdominally Based Autologous Reconstruction Methods. Plast Reconstr Surg. 2016 Mar;137(3):758–771. https://doi.org/10.1097/01.prs.0000479932.11170.8f; Pusic AL, Matros E, Fine N, Buchel E, Gordillo GM, Hamill JB, et al. Patient-Reported Outcomes 1 Year After Immediate Breast Reconstruction: Results of the Mastectomy Reconstruction Outcomes Consortium Study. J Clin Oncol. 2017 Aug 1;35(22):2499–2506. https://doi.org/10.1200/JCO.2016.69.9561; https://www.rpmj.ru/rpmj/article/view/702
-
12Academic Journal
المؤلفون: Shilov, V N, Semina, O V, Khakimova, G A, Minnebaev, D F, Akhmadullin, R M
المصدر: IOP Conference Series: Earth and Environmental Science ; volume 852, issue 1, page 012096 ; ISSN 1755-1307 1755-1315
-
13Academic Journal
المؤلفون: Khakimova, G V, Asfandiyarova, L R
المصدر: IOP Conference Series: Earth and Environmental Science ; volume 848, issue 1, page 012138 ; ISSN 1755-1307 1755-1315
-
14Academic Journal
المؤلفون: Asfandiyarova, L R, Khakimova, G V, Islamutdinova, A A
المصدر: IOP Conference Series: Earth and Environmental Science ; volume 839, issue 4, page 042093 ; ISSN 1755-1307 1755-1315
-
15Academic Journal
المؤلفون: Khakimova, G., Tryakin, A., Zabotina, T.
المصدر: Annals of Oncology ; volume 32, page S1461 ; ISSN 0923-7534
-
16Academic Journal
المؤلفون: Shtenikova, I. A., Khakimova, G. N., Zaripova, T. V., Sazhina, M. G., Штеникова, И. А., Хакимова, Г. Н., Зарипова, Т. В., Сажина, М. Г.
المصدر: Сборник статей
مصطلحات موضوعية: HISTORY OF HEALTH CARE OF THE URAL, DEVELOPMENT OF PEDIATRICS, ИСТОРИЯ ЗДРАВООХРАНЕНИЯ УРАЛА, РАЗВИТИЕ ПЕДИАТРИИ
وصف الملف: application/pdf
Relation: Сборник статей "V Международная (75 Всероссийская) научно-практическая конференция "Актуальные вопросы современной медицинской науки и здравоохранения". 2020. №2; Особенности педиатрической помощи в Свердловской области в довоенное время (1917-1941) / И. А. Штеникова, Г. Н. Хакимова, Т. В. Зарипова, М. Г. Сажина // Актуальные вопросы современной медицинской науки и здравоохранения : сборник статей V Международной (75 Всероссийской) научно-практической конференции. – 2020. – №2. – С.470-475.; http://elib.usma.ru/handle/usma/3138
الاتاحة: http://elib.usma.ru/handle/usma/3138
-
17Academic Journal
المؤلفون: O’GLI, T. A. (TASHMAMAT), KHAKIMOVA, G. (GULSHAN)
المصدر: International Journal of Innovations in Engineering Research and Technology
مصطلحات موضوعية: Multi-year training of young wrestlers, training, practice, special and general features, Indonesia
وصف الملف: application/pdf
Relation: https://www.neliti.com/publications/337101/characteristics-of-technical-training-of-young-wrestlers
-
18Academic Journal
المؤلفون: Khakimova, G.
المصدر: Annals of Oncology ; volume 31, page S1423 ; ISSN 0923-7534
-
19Academic Journal
المؤلفون: G. Khakimova G., Zh. Cherkasova R., S. Tsurkan A., G. Fedchikov A., N. Suganov V., V. Gorbunova A., Г. Хакимова Г., Ж. Черкасова Р., С. Цуркан А., Г. Федчиков А., Н. Суганов В., В. Горбунова А.
المصدر: Siberian journal of oncology; Том 18, № 5 (2019); 18-28 ; Сибирский онкологический журнал; Том 18, № 5 (2019); 18-28 ; 2312-3168 ; 1814-4861 ; 10.21294/1814-4861-2019-18-5
مصطلحات موضوعية: chemotherapy, gastric cancer, small-cell lung cancer, colorectal cancer, neuroendocrine cancer, ovarian cancer, СА-62 cancer antigen, immunochemiluminescent assay, cancer marker, monitoring, stable disease, disease progression, химиотерапия, рак желудка, мелкоклеточный рак легкого, колоректальный рак, рак яичников, нейроэндокринные опухоли, раковый антиген СА-62, иммунохемилюминесцентный анализ, онкомаркер, мониторинг, стабилизация, прогрессирование
وصف الملف: application/pdf
Relation: https://www.siboncoj.ru/jour/article/view/1184/663; Jackson S.E., Chester J.D. Personalised cancer medicine. Int J Cancer. 2015 Jul 15; 137(2): 262–6. doi:10.1002/ijc.28940.; Mascaux C., Tomasini P., Greiller I., Barilsi F. Personalised medicine for nonsmall cell lung cancer. Eur Respir Rev. 2017 Nov 15; 26(146). pii: 170066. doi:10.1183/16000617.0066-2017.; Baxendale S., van Eedan F., Wilkinson R. The power of Zebrafish in Personalised Medicine. Adv Exp Med Biol. 2017; 1007: 179–197. doi:10.1007/978-3-319-60733-7_10.; Ballman K.V. Biomarker: Predictive or Prognostic? J Clin Oncol. 2015 Nov 20; 33(33): 3968–71. doi:10.1200/JCO.2015.63.3651.; Huang J., Hu W., Sood A.K. Prognostic Biomarkers in Ovarian Cancer. Cancer Biomark. 2010–2011; 8(0): 231–251. doi:10.3233/CBM2011-0212.; Weigel M.T., Dowsett M. Current and emerging biomarkers in breast cancer: prognostic and predictive. Endocr Relat Cancer. 2010 Sep 23; 17(4): R245–62. doi:10.1677/ERC-10-0136.; Das V., Kalita J., Pal M. Predictive and prognostic biomarkers in colorectal cancer: a systematic review of recent advances and challenges. Biomed Pharmacother. 2017 Mar; 87: 8–19. doi:10.1016/j.biopha.2016.12.064.; Rodriguez-Vida A., Strijbos M., Hutson T. Predictive and prognostic biomarkers of targeted agents and modern immunotherapy in renal cell carcinoma. ESMO Open. 2016 May 25; 1(3): e000013. doi:10.1136/esmoopen-2015-000013.; Ledermann J.A., Raja F.A., Fotopoulou C., Gonzalez-Martin A., Colombo N., Sessa C.; ESMO Guidelines Working Group. Newly diagnosed and relapsed epithelial ovarian carcinoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2013 Oct; 24 Suppl 6: vi24–32. doi:10.1093/annonc/mdt333.; Parker C., Gillessen S., Heidenreich A., Horwich A.; ESMO Guidelines Committee. ESMO clinical guidelines for diagnosis, treatment and follow-up for prostate cancer. Ann Oncol. 2015 Sep; 26 Suppl 5: v69–77. doi:10.1093/annonc/mdv222.; Minimum clinical recommendations ESMO [Internet]. URL: https://www.jastro.or.jp/customer/guideline/2016/10/esmo_index.pdf (cited 10.01.2019).; Семёнова А.И. Мониторинг эффективности лечения и выявление рецидивов с помощью биомаркеров. Практическая онкология. 2011; 12(4): 171–177.; Moro R., Tcherkassova J., Song E., Shen G., Moro R., Schmid R., Hu X., Kummer A., Chen C. A new broad-spectrum cancer marker. In-Vitro Diagnostic Technology. 2005; 1–6.; Moro R., Gulyaeva-Tcherkassova J., Stieber P. Icreased AFPReceptor (RECAF) values in the serum of patients with early stages of breast cancer. J Curr Oncol. 2012; 19(1): e1–8. doi:10.3747/co.19.979.; Tcherkassova J., Abramovich C., Moro R., Chen C., Smit R., Gerber A., Moro R. Combination of CA125 and RECAF biomarkers for early detection of Ovarian cancer. Tumor Biol. 2011; 32(4): 831–838. doi:10.1007/s13277-011-0186-1.; Moro R., Uriel J. Early localization of AFP in the developing nervous system of the chicken. Oncodevelop Biol Med. 1981; 2: 391–398.; Villacampa M.J., Moro R., Naval J., Failly-Crepin C. Lampreave F., Uriel J. RECAFs in a human breast cancer cell line. Biochem Biophys Res Commun. 1984; 122: 1322–1327. doi:10.1016/0006-291X(84)91236-1.; Jacobson H.I., Bennett J.A., Mizejewski G.J. Inhibition of estrogendependent breast cancer growth by a reaction product of alpha-fetoprotein and estradiol. Cancer Res. 1990; 50(2): 415–420.; Mizejewski G.J., Phillips L., Stoll W. In vitro studies of the abortogenic potential of anti-serum to alpha-fetoprotein. Int J Immunopharmacol. 1981; 3(1): 87–95.; Moro R., Tamaoki T., Wegmann T.G., Longenecker B.M., Laderoute M.P. Monoclonal antibodies directed against a widespread oncofetal antigen: the alpha-feloprotein receptor. Tumour Biol. 1993; 14(2): 116–30. doi:10.1159/000217864.; Laderoute M. A new perspective on the nature of the cancer problem: anticellular senescence. Mol Carcinog. 1994 Jul; 10(3): 125–33.; Uriel J., Villacampa M.J., Moro R., Naval l., Failly-Crepin C. Uptake of radiolabeled alpha•fetoprolein by mouse mammary carcinomas and its usefulness in tumor scintigraphy. Cancer Res. 1984; 44: 5314–5319.; Pepe M.S., Etzioni R., Feng Z., Potter J.D., Thompson M.L., Thornquist M., Winget M., Yasui Y. Phases of biomarker development for early detection of cancer. J Natl Cancer Inst. 2001 Jul 18; 93(14): 1054–61. doi:10.1093/jnci/93.14.1054.; Uriel J., Poupon M.F., Geuskens M. Alphafoecoprolein uptake by cloned cell lines derived from a nickel•induced rat rhabdomyosarcoma. J Cancer. 1983; 48: 261–269.; Suzuki Y., Zeng C.Q., Alpert E. Isolation and partial characterization of a specific alpha-fetoprotein receptor on human monocytes. Clin Invest. 1992; 90: 1530–6. doi:10.1172/JCI116021.; Torres J.M., Darracq N., Uriel J. Membrane proteins from Lymphoblastoid cells showing cross-affinity for alpha-fetoprotein and albumin. Isolation and characterization. Biochim Biophys Acta. 1992 Sep 4; 1159(1): 60–6.; Kanevsky V.Yu., Pozdnyakova L.P., Aksenova O.A., Severin S.E., Katukov V.Yu., Severin E.S. Isolation and characterization of AFP binding proteins from tumor and fetal human tissues. Biochem Mol Biol Int. 1997; 41(6): 1143–51.; Черешнев В.А., Родионов С.Ю., Васильев Н.В., Орлов О.А., Черкасов В.А. Иммунотерапия злокачественных опухолей альфафетопротеином человека как этап комбинированного и комплексного лечения онкологических больных [Интернет]. URL: http://www.a-fp.ru/articles/immunoterapiya-zlokachestvennyh-opuholey-alfa-fetoproteinomcheloveka-kak-etap (дата обращения: 10.01.2019).; Tcherkassova J., Tsurkan S., Smirnova G., Borisova J., Moro R., Treshalina H. Binding characterization of the targetingdrug AIMPILA to AFP receptors in human tumor xenografts. Tumour Biol. 2017 Oct; 39(10): 1010428317734815. doi:10.1177/1010428317734815.; Ohkawa K., Tsukada Y., Abe T., Takada K., Hibi N. Overcoming effect of antibody against rat alpha-fetoprotein (AFP) on the growth of daunorubicin-resistant mutant rat ascites hepatoma cell line AH66. Int J Cancer. 1989; 44(3): 489–493. doi:10.1002/ijc.2910440319.; Астахов Д.В., Коваленко Н.А., Сологуб В.К., Большакова Т.Д., Северин С.Е., Шатверян Г.А., Гудовский Л.М. Определение антител к рецептору альфа-фетопротеина у онкологических больных. Вопросы биологической, медицинской и фармакологической химии. 1999; 2: 27–30.; Molina R., Auge J.M., Escudero J.M., Marrades R., Viñolas N., Carcereny E., Ramirez J., Filella X. Mucins CA 125, CA 19-9, CA 15.3, and TAG 72.3 as tumor markers in patinets with lung cancer: comparison with CYFRA 21-1, CEA, SCC and NSE. Tumour Biol. 2008; 29(6): 371–80. doi:10.1159/000181180.; Molina R., Augé J.M., Bosch X., Escudero J.M., Viñolas N., Marrades R., Ramírez J., Carcereny E., Filella X. Usefulness of serum tumor markers, including progastrin-releasing peptide in patients with lung cancer. Correlation and histology. Tumour Biol. 2009; 30(3): 121–9. doi:10.1159/000224628.; European Group on Tumor markers. Consensus recommendations. Anticancer Res. 1999, 19: 2785–820.; Iwanicki-Caron I., Di Fiore F., Roque I., Astruc E., Stetiu M., Duclos A., Tougeron D., Saillard S., Thureau S., Benichou J., Paillot B., Basuyau J.P., Michel P. Usefulness of the serum carcinoembryonic antigen kinetic for chemotherapy monitoring in patients with unresectable metastasis of colorectal cancer. J Clin Oncol. 2008 Aug 1; 26(22): 3681–6. doi:10.1200/JCO.2007.15.0904.; Li C.S., Cheng B.C., Ge W., Gao J.F. Clinical value of CYFRA21-1, NSE, CA15-3, CA19-9 and CA125 assay in the elderly patients with pleural effusions. nt J Clin Pract. 2007 Mar; 61(3): 444–8. doi:10.1111/j.17421241.2006.01111.x.; Prados M.C., Alvarez-Sala R., Blasco R., Chivato T., García Satué J.L., García Río F.J., Gómez de Terrero F.J. Villamor The clinical value of neuron-specific enolase as a tumor marker in bronchoalveolar lavage. Cancer. 1994 Sep 1; 74(5): 1552–5. doi:10.1002/1097-0142(19940901)74:53.0.co;2-f.; Karnak D., Beder S., Kayacan O., Ibis E., Oflaz G. Neuron-specific enolase and lung cancer. Am J Clin Oncol. 2005 Dec; 28(6): 586–90.; Saba M.A., Valeh T., Ehteram H., Haddad Kashani H., Ghazi Zahedi M. Diagnostic Value of Neuron-Specific Enolase (NSE) and Cancer Antigen 15-3 (CA 15-3) in the Diagnosis of Pleural Effusions. Asian Pac J Cancer Prev. 2017 Jan 1; 18(1): 257–261. doi:10.22034/APJCP.2017.18.1.257.; Jørgensen L.G., Osterlind K., Genollá J., Gomm S.A., Hernández J.R., Johnson P.W., Løber J., Splinter T.A., Szturmowicz M. Serum neuron-specific enolase (S-NSE) and the prognosis in small cell lung cancer (SCLC): a combined multivariable analysis on data from nine centers. Br. J. Cancer 1996; 74; 463–467. doi:10.1038/bjc.1996.383.; Carney D.N., Teeling M. Neuron-specific enolase: how useful as a cancer marker? Eur J Cancer Clin Oncol. 1988 May; 24(5): 825–8.; https://www.siboncoj.ru/jour/article/view/1184
-
20Academic Journal
المؤلفون: G. Khakimova G., A. Tryakin A., T. Zabotina N., A. Tsukanov S., V. Aliev A., S. Gutorov L., Г. Хакимова Г., А. Трякин А., Т. Заботина Н., А. Цуканов С., В. Алиев А., С. Гуторов Л.
المصدر: Malignant tumours; Том 9, № 2 (2019); 5-11 ; Злокачественные опухоли; Том 9, № 2 (2019); 5-11 ; 2587-6813 ; 2224-5057
مصطلحات موضوعية: intestinal microbiome, immunotherapy, CTLA-4 PD-1, кишечная микробиота, иммунотерапия
وصف الملف: application/pdf
Relation: https://www.malignanttumors.org/jour/article/view/596/434; Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, JemalA. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65 (2):87-108.; Fearon ER. Molecular genetics of colorectal cancer. Ann Rev Pathol. 2011;6 (1):479-507.; Jafri SH, Mills G. Lifestyle modification in colorectal cancer patients: an integrative oncology approach. Future Oncol. 2013;9 (2):207-218.; Okugawa Y, Grady WM, Goel A. Epigenetic alterations in colorectal cancer: emerging biomarkers. Gastroenterology. 2015;149 (5):1204-1225.; Lasry A, Zinger A, Ben-Neriah Y. Inflammatory networks underlying colorectal cancer. Nat Immunol. 2016;17 (3):230-240.; O'Keefe SJ. Diet, microorganisms and their metabolites, and colon cancer. Nat Rev Gastroenterol Hepatol. 2016;13 (12):691-706.; Jess T, Simonsen J, Jcrgensen KT, Pedersen BV, Nielsen NM, Frisch M. Decreasing risk of colorectal cancer in patients with inflammatory bowel disease over 30 years. Gastroenterology. 2012;143 (2):375-381.; Jess T, Horvath-Puho E, Fallingborg J, Rasmussen HH, Jacobsen BA. Cancer risk in inflammatory bowel disease according to patient phenotype and treatment: a Danish population-based cohort study. Am J Gastroenterol. 2013;108 (12):1869-1876.; Johnson CM, Wei C, Ensor JE, et al. Meta-analyses of colorectal cancer risk factors. Cancer Causes Control. 2013;24 (6):1207-1222.; Farraye FA, Odze RD, Eaden J, Itzkowitz SH. AGA medical position statement on the diagnosis and management of colorectal neoplasia in inflammatory bowel disease. Gastroenterology. 2010;138 (2):738-745.; Kostic AD, Chun E, Meyerson M, Garrett WS. Microbes and inflammation in colorectal cancer. Cancer Immunol Res. 2013;1 (3):150-157.; Bruner SD, Jobin C. Intestinal microbiota in inflammatory bowel disease and carcinogenesis: implication for therapeutics. Clin Pharmacol Ther. 2016;99 (6):585-587.; Ijssennagger N, van der Meer R, van Mil SWC. Sulfide as a mucus barrier-breaker in inflammatory bowel disease? Trends Mol Med. 2016; 22 (3):190-199.; Uronis JM, Muhlbauer M, Herfarth HH, Rubinas TC, Jones GS, Jobin C. Modulation of the intestinal microbiota alters colitis-associated colorectal cancer susceptibility. PLoS One. 2009;4 (6):e6026.; Li Y, Kundu P, Seow SW, et al. Gut microbiota accelerate tumor growth via c-jun and STAT3 phosphorylation in APC Min/ + mice. Carcinogenesis. 2012;33 (6):1231—1238.; Васильев А.Н. Трансплантация фекальной микробиоты: возможные терапевтические подходы и вопросы правового регулирования / А.Н. Васильев, Д.В. Горячев, Е.В. Гавришина и др. // Биопрепараты. Рецензируемый научно-практический журнал. 2015. No 2 (54) С. 15-23.; Yu J, Feng Q, Wong SH, et al. Metagenomic analysis of faecal microbiome as a tool towards targeted non-invasive biomarkers for colorectal cancer. Gut. 2017;66 (1):70-78.; Crawford PA, Gordon JI. From the cover: microbial regulation of intestinal radiosensitivity. Proc Natl Acad Sci. 2005;102 (37): 13254-13259.; Yu T, Guo F, Yu Y, et al. Fusobacterium nucleatum promotes chemoresistance to colorectal cancer by modulating autophagy. Cell. 2017; 170 (3):548-563.; Vetizou M, Pitt JM, Daillere R, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015; 350 (6264):1079-1084.; Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences efficacy of PD-1 based immunotherapy against epithelial tumors. Science. 2018;359 (6371):91-97.; Ahmed S, Johnson K, Ahmed O, Iqbal N. Advances in the management of colorectal cancer: from biology to treatment. Int J Colorectal Dis. 2014;29 (9):1031-1042.; Field K, Lipton L. Metastatic colorectal cancer-past, progress and future. World J Gastroenterol. 2007;13 (28):3806-3815.; Dejea CM, Wick EC, Hechenbleikner EM, et al. Microbiota organization is a distinct feature of proximal colorectal cancers. Proc Natl Acad Sci. 2014;111 (51):18321-18326.; Li S, Konstantinov SR, Smits R, Peppelenbosch MP. Bacterial biofilms in colorectal cancer initiation and progression. Trends Mol Med. 2017; 23 (1):18-30.; Sicard J-F, Le Bihan G, Vogeleer P, Jacques M, Harel J. Interactions of intestinal bacteria with components of the intestinal mucus. Front Cell Infect Microbiol. 2017 ;7:387.; Nakatsu G, Li X, Zhou H, et al. Gut mucosal microbiome across stages of colorectal carcinogenesis. Nat Commun. 2015;6 (1):8727.; Huipeng W, Lifeng G, Chuang G, Jiaying Z, Yuankun C. The differences in colonic mucosal microbiota between normal individual and colon cancer patients by polymerase chain reaction-denaturing gradient gel electrophoresis. J Clin Gastroenterol. 2014;48 (2):138-144.; Warren RL, Freeman DJ, Pleasance S, et al. Co-occurrence of anaerobic bacteria in colorectal carcinomas. Microbiome. 2013;1 (1):16.; Wu N, Yang X, Zhang R, et al. Dysbiosis signature of fecal microbiota in colorectal cancer patients. Microb Ecol. 2013;66 (2):462-470.; Kahouli I, Tomaro-Duchesneau C, Prakash S. Probiotics in colorectal cancer (CRC) with emphasis on mechanisms of action and current perspectives. J Med Microbiol. 2013;62 (Pt_8):1107-1123.; Ito M, Kanno S, Nosho K, et al. Association of Fusobacterium nucleatum with clinical and molecular features in colorectal serrated pathway. Int J Cancer. 2015;137 (6):1258-1268.; Park CH, Han DS, Oh Y-H, Lee A-Reum, Lee Y-Ra, Eun CS. Role of Fusobacteria in the serrated pathway of colorectal carcinogenesis. Sci Rep. 2016;6 (1):25271.; Ye X, Wang R, Bhattacharya R, et al. Fusobacterium nucleatum subspecies Animalis influences proinflammatory cytokine expression and monocyte activation in human colorectal tumors. Cancer Prev Res. 2017;10 (7):398-409.; Nosho K, Sukawa Y, Adachi Y, et al. Association of Fusobacterium nucleatum with immunity and molecular alterations in colorectal cancer. World J Gastroenterol. 2016;22 (2):557-566.; de Vries NL, Swets M, Vahrmeijer AL, Hokland M, Kuppen PJ. The immunogenicity of colorectal cancer in relation to tumor development and treatment. Int J Mol Sci. 2016;17 (7):E1030.; Nishino M, Ramaiya NH, Hatabu H, Hodi FS. Monitoring immunecheckpoint blockade: response evaluation and biomarker development. Nat Rev Clin Oncol. 2017;14 (11):655—668.; Stewart CA, Metheny H, Iida N, et al. Interferon-dependent IL-10 production by Tregs limits tumor Th17 inflammation. J Clin Invest. 2013;123 (11):4859-4874.; Pang Y, Gara SK, Achyut BR, et al. TGF-P signaling in myeloid cells is required for tumor metastasis. Cancer Discov. 2013;3 (8):936-951.; Qian X, Chen H, Wu X, Hu L, Huang Q, Jin Y. Interleukin-17 acts as double-edged sword in anti-tumor immunity and tumorigenesis. Cytokine. 2017;89:34-44.; Pitt JM, Vetizou M, Waldschmitt N, et al. Fine-tuning cancer immunotherapy: optimizing the gut microbiome. Cancer Res. 2016; 76 (16):4602-4607.; West NR, Powrie F. Immunotherapy not working? Check your microbiota. Cancer Cell. 2015;28 (6):687-689.; Snyder A, Pamer E, Wolchok J. Immunotherapy. Could microbial therapy boost cancer immunotherapy? Science. 2015;350 (6264):1031-1032.; Tanaka A, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Cell Res. 2017;27 (1):109-118.; Geis AL, Fan H, Wu X, et al. Regulatory T-cell response to entero-toxigenic Bacteroides fragilis colonization triggers IL17-dependent colon carcinogenesis. Cancer Discov. 2015;5 (10):1098-1109.; Viaud S, Saccheri F, Mignot G, et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science. 2013;342 (6161):971-976.; Baecher-Allan C, Viglietta V, Hafler DA. Human CD4+CD25+ regulatory T cells. Semin Immunol. 2004;16 (2):89-98.; Egen JG, Kuhns MS, Allison JP. CTLA-4: new insights into its biological function and use in tumor immunotherapy. Nat Immunol. 2002; 3 (7):611-618.; Dubin K, Callahan MK, Ren B, et al. Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat Commun. 2016;7:10391.; Nishino M, Ramaiya NH, Hatabu H, Hodi FS. Monitoring immunecheckpoint blockade: response evaluation and biomarker development. Nat Rev Clin Oncol. 2017;14 (11):655-668.; Gopalakrishnan V, Spencer CN, Nezi L, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359 (6371):97-103.; Sivan A, Corrales L, Hubert N, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350 (6264):1084-1089.; Matson V, Fessler J, Bao R, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359 (6371):104-108.; Jin P, Wang K, Huang C, Nice EC. Mining the fecal proteome: from biomarkers to personalised medicine. Expert Rev Proteomics. 2017; 14 (5):445-459.; Ai L, Tian H, Chen Z, Chen H, Xu J, Fang JY. Systematic evaluation of supervised classifiers for fecal microbiota-based prediction of colorectal cancer. Oncotarget. 2017;8 (6):9546-9556.; Zhang MM, Cheng JQ, Xia L, et al. Monitoring intestinal microbiota profile: a promising method for the ultraearly detection of colorectal cancer. Med Hypotheses. 2011;76 (5):670-672.; Shah MS, Desantis TZ, Weinmaier T, et al. Leveraging sequencebased faecal microbial community survey data to identify a composite biomarker for colorectal cancer. Gut. 2018;67 (5):882-891.; Enq RR. How's your microbiota? Let's check your urine. Blood. 2015; 126 (14):1641-1642.; Ferreira RM, Pereira-Marques J, Pinto-Ribeiro I, et al. Gastric microbial community profiling reveals a dysbiotic cancer-associated microbiota. Gut. 2018;67 (2):226-236.; Panteli JT, Forkus BA, van Dessel N, Forbes NS. Genetically modified bacteria as a tool to detect microscopic solid tumor masses with triggered release of a recombinant biomarker. Integr Biol. 2015;7 (4):423-434.; Gardlik R, Fruehauf JH. Bacterial vectors and delivery systems in cancer therapy. IDrugs. 010;13(10):701-706.; https://www.malignanttumors.org/jour/article/view/596