يعرض 1 - 10 نتائج من 1,136 نتيجة بحث عن '"Makatsariya, A"', وقت الاستعلام: 1.06s تنقيح النتائج
  1. 1
    دورية أكاديمية

    المصدر: Ultrasound in obstetrics & gynecology : the official journal of the International Society of Ultrasound in Obstetrics and Gynecology. 55(4):441-449

    مصطلحات موضوعية: Medicin och hälsovetenskap

  2. 2
    دورية أكاديمية
  3. 3
    دورية أكاديمية

    المساهمون: The work was financially supported by Sanofi pharmaceutical company, Работа выполнена при финансовой поддержке фармацевтической компании Санофи

    المصدر: Obstetrics, Gynecology and Reproduction; Vol 18, No 2 (2024); 218-230 ; Акушерство, Гинекология и Репродукция; Vol 18, No 2 (2024); 218-230 ; 2500-3194 ; 2313-7347

    وصف الملف: application/pdf

    العلاقة: https://www.gynecology.su/jour/article/view/2053/1202Test; Grober U., Schmidt J., Kisters K. Magnesium in prevention and therapy. Nutrients. 2015;7(9):8199–226. doi:10.3390/nu7095388.; Bertinato J., Wu Xiao C., Ratnayake W.M. et al. Lower serum magnesium concentration is associated with diabetes, insulin resistance, and obesity in South Asian and white Canadian women but not men. Food Nutr Res. 2015;59(1):25974. doi:10.3402/fnr.v59.25974.; Al Alawi M.A., Majoni S.W., Falhammar H. Magnesium and human health: perspectives and research directions. Int J Endocrinol. 2018;2018:9041694. doi:10.1155/2018/9041694.; Громова О.А., Торшин И.Ю., Рудаков К.В. и др. Недостаточность магния – достоверный фактор риска коморбидных состояний: результаты крупномасштабного скрининга магниевого статуса в регионах России. Фарматека. 2013;(6):115–29.; Блинов Д.В., Ушакова Т.И., Макацария Н.А. и др. Гормональная контрацепция и дефицит магния: результаты субанализа исследования MAGYN. Акушерство, Гинекология и Репродукция. 2017;11(1):36–48. doi:10.17749/2313-7347.2017.11.1.036-048.; Блинов Д.В., Зимовина У.В., Джобава Э.М. Ведение беременных с дефицитом магния: фармакоэпидемиологическое исследование. ФАРМАКОЭКОНОМИКА. Современная фармакоэкономика и фармакоэпидемиология. 2014;7(2):23–32.; Makatsariya A.D., Bitsadze V.O, Blinov D.V. et al. Pregnant women with symptoms of magnesium deficiency in Russian Federation: MAGIC 2 study results. Magnes Res. 2016;29(3):81. URL: https://www.researchgate.net/publication/316007448_Pregnant_women_with_symptoms_of_magnesium_deficiency_in_Russian_Federation_MAGIC_2_study_resultsTest.; Блинов Д.В., Зимовина У.В., Сандакова Е.А., Ушакова Т.И. Дефицит магния у пациенток с гормонально-зависимыми заболеваниями: фармакоэпидемиологический профиль и оценка качества жизни. ФАРМАКОЭКОНОМИКА. Современная фармакоэкономика и фармакоэпидемиология. 2015;8(2):16–24. URL: https://cyberleninka.ru/article/n/defitsit-magniya-u-patsientok-s-gormonalno-zavisimymi-zabolevaniyami-farmakoepidemiologicheskiy-profil-i-otsenka-kachestva-zhizniTest.; Schimatschek H.F., Rempis R. Prevalence of hypomagnesemia in an unselected German population of 16,000 individuals. Magnes Res. 2001;14(4):283–90.; De Baaij J.H.F., Hoenderop J.G.J., Bindels R.J.M. Magnesium in man: implications for health and disease. Physiol Rev. 2015;95(1):1–46. doi:10.1152/physrev.00012.2014.; Olza J., Aranceta-Bartrina J., González-Gross M. et al. Reported dietary intake, disparity between the reported consumption and the level needed for adequacy and food sources of calcium, phosphorus, magnesium and vitamin D in the Spanish population: findings from the ANIBES study. Nutrients. 2017;9(2):168. doi:10.3390/nu9020168.; Olza J., Aranceta-Bartrina J., González-Gross M. et al. Reported dietary intake and food sources of zinc, selenium, and vitamins A, E and C in the Spanish population: findings from the ANIBES study. Nutrients. 2017;9(7):697. doi:10.3390/nu9070697.; Jahnen-Dechent W., Ketteler M. Magnesium basics. Clin Kidney J. 2012;5(Suppl 1):i3–i14. doi:10.1093/ndtplus/sfr163.; Danziger J., William J.H., Scott D.J. et al. Proton-pump inhibitor use is associated with low serum magnesium concentrations. Kidney Int. 2013;83(4):692–9. doi:10.1038/ki.2012.452.; Наумов Д.Е. Термочувствительные ионные каналы TRPM8 (oбзор литературы). Бюллетень физиологии и патологии дыхания. 2011;(42):89–96.; Громова О.А., Гоголева И.В. Применение магния в зеркале доказательной медицины и фундаментальных исследований в терапии. Дефицит магния и концепция стресса. Трудный пациент. 2007;5(11):29–38.; Voets T., Nilius B., Hoefs S. et al. TRPM6 Forms the Mg 2+ influx channel involved in intestinal and renal Mg 2+ absorption. J Biol Chem. 2004;279(1):19–25. doi:10.1074/jbc.M311201200.; Schlingmann K.P., Gudermann T. A critical role of TRPM channel-kinase for human magnesium transport. J Physiol. 2005;566(Pt 2):301–8. doi:10.1113/jphysiol.2004.080200.; Pilchova I., Klacanova K., Tatarkova Z. et al. The involvement of Mg 2+ in regulation of cellular and mitochondrial functions. Oxid Med Cell Longev. 2017;2017:6797460. doi:10.1155/2017/6797460.; Sontia B., Touyz R.M. Magnesium transport in hypertension. Pathophysiology. 2007;14(3–4):205–11. doi:10.1016/j.pathophys.2007.09.005.; Mutnuri S., Fernandez I., Kochar T. Suppression of parathyroid hormone in a patient with severe magnesium depletion. Case Rep Nephrol. 2016;2016:2608538. doi:10.1155/2016/2608538.; Viering D.H.H.M., de Baaij J.H.F., Walsh S.B. et al. Genetic causes of hypomagnesemia, a clinical overview. Pediatr Nephrol. 2017;32(7):1123–35. doi:10.1007/s00467-016-3416-3.; Seo J.W., Park T.J. Magnesium metabolism. Electrolyte Blood Press. 2008;6(2):86–95. doi:10.5049/EBP.2008.6.2.86.; Pham P.C.T., Pham P.-A.T., Pham S.V. et al. Hypomagnesemia: a clinical perspective. Int J Nephrol Renovasc Dis. 2014;7:219–30. doi:10.2147/IJNRD.S42054.; Lajer H., Daugaard G. Cisplatin and hypomagnesemia. Cancer Treat Rev. 1999;25(1):47–58. doi:10.1053/ctrv.1999.0097.; Bagnis C.I., Deray G. Amphotericin B nephrotoxicity. Saudi J Kidney Dis Transpl. 2002;13(4):481–91.; Lee C.H., Kim G.-H. Electrolyte and acid-base disturbances induced by clacineurin inhibitors. Electrolyte Blood Press. 2007;5(2):126–30. doi:10.5049/EBP.2007.5.2.126.; Sivakumar J. Proton pump inhibitor-induced hypomagnesaemia and hypocalcaemia : case review. Int J Physiol Pathophysiol Pharmacol. 2016;8(4):169–74.; Atsmon J., Dolev E. Drug-induced hypomagnesaemia: scope and management. Drug Saf. 2005;28(9):763–88. doi:10.2165/00002018-200528090-00003.; Makatsariya A.D., Dzhobava E.M., Bitsadze V.O. et al. Observational study of outpatient women in hormone dependent conditions with magnesium deficiency and receiving Magne B6® Forte in Russia (MAGYN Study). Magnes Res. 2016;29(3):82. URL: https://www.researchgate.net/publication/316007423_Observational_study_of_outpatient_women_in_hormone_dependent_conditions_with_magnesium_deficiency_and_receiving_Magne_B6_Forte_in_Russia_MAGYN_StudyTest.; Ryu A., Kim T.H. Premenstrual syndrome : a mini review. Maturitas. 2015;82(4):436–40. doi:10.1016/j.maturitas.2015.08.010.; Rosenstein D.L., Elin R.J., Hosseini J.M. et al. Magnesium measures across the menstrual cycle in premenstrual syndrome. Biol Psychiatry. 1994;35(8):557–61. doi:10.1016/0006-3223(94)90103-1.; Sherwood R.A., Rocks B.F., Stewart A., Saxton R.S. Magnesium and the premenstrual syndrome. Ann Clin Biochem. 1986;23(Pt 6):667–70. doi:10.1177/000456328602300607.; Muneyyirci-Delale O., Nacharaju V.L., Altura B.M., Altura B.T. Sex steroid hormones modulate serum ionized magnesium and calcium levels throughout the menstrual cycle in women. Fertil Steril. 1998;69(5):958–2. doi:10.1016/S0015-0282(98)00053-3.; Tonick S., Muneyyirci-Delale O. Magnesium in women’s health and gynecology. Open J Obstet Gynecol. 2016;6(5):325–33. doi:10.4236/ojog.2016.65041.; Facchinetti F., Sances G., Borella P. et al. Magnesium prophylaxis of menstrual migraine: effects on intracellular magnesium. Headache. 1991;31(5):298–301. doi:10.1111/j.1526-4610.1991.hed3105298.x.; Walker A.F., De Souza M.C., Vickers M.F. et al. Magnesium supplementation alleviates premenstrual symptoms of fluid retention. J Womens Health. 1998;7(9):1157–65. doi:10.1089/jwh.1998.7.1157.; Quaranta S., Buscaglia M.A., Meroni M.G. et al. Pilot study of the efficacy and safety of a modified-release magnesium 250 mg tablet (Sincromag) for the treatment of premenstrual syndrome. Clin Drug Investig. 2007;27(1):51–8. doi:10.2165/00044011-200727010-00004.; De Souza M.C., Walker A.F., Robinson P.A., Bolland K. A synergistic effect of a daily supplement for 1 month of 200 mg magnesium plus 50 mg vitamin B6 for the relief of anxiety-related premenstrual symptoms: a randomized, double-blind, crossover study. J Womens Health Gend Based Med. 2000;9(2):131–9. doi:10.1089/152460900318623.; Fathizadeh N., Ebrahimi E., Valiani M. et al. Evaluating the effect of magnesium and magnesium plus vitamin B6 supplement on the severity of premenstrual syndrome. Iran J Nurs Midwifery Res. 2010;15(Suppl 1):401–5.; Дадак К., Макацария А.Д., Блинов Д.В., Зимовина У.В. Клинические и биохимические аспекты применения препаратов магния в акушерстве, гинекологии и перинатологии. Акушерство, Гинекология и Репродукция. 2014;8(2):69–78.; Черкасова Н.Ю., Фомина А.В., Филиппова О.В. Анализ рынка лекарственных средств для лечения дисменореи. ФАРМАКОЭКОНОМИКА. Современная фармакоэкономика и фармакоэпидемиология. 2013;6(3):36–9.; Унанян А.Л., Алимов В.А., Аракелов С.Э. и др. Фармакоэпидемиология использования оригинального дротаверина при дисменорее: результаты международного многоцентрового исследования. ФАРМАКОЭКОНОМИКА. Современная фармакоэкономика и фармакоэпидемиология. 2014;7(3):44–50.; Оразов М.Р., Чайка А.В., Носенко Е.Н. Купирование хронической тазовой боли, обусловленной аденомиозом, прогестагенами нового поколения. Акушерство, Гинекология и Репродукция. 2014;8(3):6–10.; Джобава Э.М. Вопросы безопасности применения дротаверина в акушерской практике. Акушерство, Гинекология и Репродукция. 2018;12(1):54–60. doi:10.17749/2313-7347.2018.12.1.054-060.; Proctor M.L., Farquhar C.M. Dysmenorrhoea. BMJ Clin Evid. 2007;2007:0813.; Proctor M., Murphy P. A. Herbal and dietary therapies for primary and secondary dysmenorrhoea. Cochrane Database Syst Rev. 2001;(3):CD002124. doi:10.1002/14651858.CD002124.; Seifert B., Wagler P., Dartsch S. et al. Magnesium – a new therapeutic alternative in primary dysmenorrhea. Zentralbl Gynakol. 1989;111(11):755–60. (In German).; Parazzini F., Di Martino M., Pellegrino P. Magnesium in the gynecological practice : a literature review. Magnes Res. 2017;30(1):1–7. doi:10.1684/mrh.2017.0419.; Fontana-Klaiber H., Hogg B. Therapeutic effects of magnesium in dysmenorrhea. Schweiz Rundsch Med Prax. 1990;79(16):491–4. (In German).; Benassi L., Barletta F.P., Baroncini L. et al. Effectiveness of magnesium pidolate in the prophylactic treatment of primary dysmenorrhea. Clin Exp Obstet Gynecol. 1992;19(3):176–9.; Higdon J. An evidence-based approach to vitamins and minerals: health benefits and intake recommendations. Stuttgart, New York: Thieme, 2012. 282 p.; Olatunji L.A., Oyeyipo I.P., Micheal O.S., Soladoye A.O. Effect of dietary magnesium on glucose tolerance and plasma lipid during oral contraceptive administration in female rats. Afr J Med Med Sci. 2008;37(2):135–9.; Akinloye O., Adebayo T.O., Oguntibeju O.O. et al. Effects of contraceptives on serum trace elements, calcium and phosphorus levels. West Indian Med J. 2011;60(3):308–15.; Muneyyirci-Delale O., Nacharaju V.L., Dalloul M. et al. Divalent cations in women with PCOS: implications for cardiovascular disease. Gynecol Endocrinol. 2001;15(3):198–201. doi:10.1080/gye.15.3.198.201.; Sharifi F., Mazloomi S., Hajihosseini R. et al. Serum magnesium concentrations in polycystic ovary syndrome and its association with insulin resistance. Gynecol Endocrinol. 2012;28(1):7–11. doi:10.3109/09513590.2011.579663.; O’Shaughnessy A., Muneyyirci-Delale O., Nacharaju V.L. et al. Circulating divalent cations in asymptomatic ovarian hyperstimulation and in vitro fertilization patients. Gynecol Obstet Invest. 2001;52(4):237–42. doi:10.1159/000052982.; Bird S.T., Hartzema A.G., Brophy J.M. et al. Risk of venous thromboembolism in women with polycystic ovary syndrome: a population-based matched cohort analysis. CMAJ. 2013;185(2):E115–20. doi:10.1503/cmaj.120677.; Громова О.А., Лиманова О.А., Торшин И.Ю. Систематический анализ фундаментальных и клинических исследований указывает на необходимость совместного использования эстроген-содержащих препаратов с препаратами пиридоксина и магния. Акушерство, Гинекология и Репродукция. 2013;7(3):35–50.; Farsinejad-Marj M., Saneei P., Esmaillzadeh A. Dietary magnesium intake, bone mineral density and risk of fracture : a systematic review and meta-analysis. Osteoporos Int. 2016;27(4):1389–99. doi:10.1007/s00198-015-3400-y.; Gur A., Colpan L., Nas K. et al. The role of trace minerals in the pathogenesis of postmenopausal osteoporosis and a new effect of calcitonin. J Bone Miner Metab. 2002;20(1):39–43. doi:10.1007/s774-002-8445-y.; Brodowski J. Levels of ionized magnesium in women with various stages of postmenopausal osteoporosis progression evaluated on the basis of densitometric examinations. Przegl Lek. 2000;57(12):714–6. (In Polish).; Дефицит магния в акушерстве и гинекологии: результаты национального совещания. Акушерство, Гинекология и Репродукция. 2014;8(2):6–10.; Солопова А.Г., Блинов Д.В., Бегович Ё. и др. Неврологические расстройства после гистерэктомии: от патогенеза к клинике. Эпилепсия и пароксизмальные состояния. 2022;14(1):54–64. doi:10.17749/2077-8333/epi.par.con.2022.115.; Блинов Д.В., Солопова А.Г., Плутницкий А.Н. и др. Организация здравоохранения в сфере реабилитации пациенток с онкологическими заболеваниями репродуктивной системы. ФАРМАКОЭКОНОМИКА. Современная фармакоэкономика и фармакоэпидемиология. 2022;15(1):119–30. URL: https://cyberleninka.ru/article/n/organizatsiya-zdravoohraneniya-v-sfere-reabilitatsii-patsientok-s-onkologicheskimi-zabolevaniyami-reproduktivnoy-sistemyTest.; Блинов Д.В., Солопова А.Г., Ачкасов Е.Е. и др. Медицинская реабилитация пациенток с климактерическим синдромом и хирургической менопаузой: вклад коррекции дефицита магния. ФАРМАКОЭКОНОМИКА. Современная фармакоэкономика и фармакоэпидемиология. 2022;15(4):478–90. doi:10.17749/2070-4909/farmakoekonomika.2022.159.; Блинов Д.В., Солопова А.Г., Ачкасов Е.Е. и др. Роль коррекции дефицита магния в реабилитации женщин с климактерическим синдромом и хирургической менопаузой: результаты исследования MAGYN. Акушерство, Гинекология и Репродукция. 2022;16(6):676–91. doi:10.17749/2313-7347/ob.gyn.rep.2022.371.; Блинов Д.В., Солопова А.Г., Ачкасов Е.Е. и др. Организация реабилитации пациенток с опухолями яичников: современные подходы и будущие направления. ФАРМАКОЭКОНОМИКА. Современная фармакоэкономика и фармакоэпидемиология. 2023;16(2):303–16. URL: https://www.elibrary.ru/ip_restricted.asp?rpage=https%3A%2F%2Fwww%2Eelibrary%2Eru%2Fitem%2Easp%3Fedn%3DdcaonyTest.; Блинов Д.В., Солопова А.Г., Ачкасов Е.Е. и др. Алгоритм комплексной психотерапевтической поддержки для женщин с психоневрологическими симптомами в период реабилитации после лечения злокачественных новообразований репродуктивной системы. Эпилепсия и пароксизмальные состояния. 2023;15(3):232–45. doi:10.17749/2077-8333/epi.par.con.2023.168.; Магне B6. Инструкция по медицинскому применению. ЛСР-007053/09. Режим доступа: http://www.grls.rosminzdrav.ruTest. [Дата доступа: 13. 01. 2024].; Магне B6 Форте. Инструкция по медицинскому применению. ЛСР-007053/09. Режим доступа: http://www.grls.rosminzdrav.ruTest]. [Дата доступа: 13. 01. 2024].; Дижевская Е.В. Мультидисциплинарный подход к коррекции магний-дефицитных состояний. Акушерство, Гинекология и Репродукция. 2015;9(3):68–85.; Дижевская Е.В. Обмен научными данными и экспертными мнениями по фармакотерапии в течение беременности: традиционные и современные подходы III Международный экспертный совет по проблемам дефицита магния в акушерстве и гинекологии. Акушерство, Гинекология и Репродукция. 2015;9(4):93–101.; О Пленуме Президиума Российского общества акушеров-гинекологов. Акушерство и гинекология. 2015;(5):113–5.; Громова О.А. Дефицит магния как проблема современного питания у детей и подростков. Педиатрическая фармакология. 2014;(1):20–30.; Ranade V.V., Somberg J.C. Bioavailability and pharmacokinetics of magnesium after administration of magnesium salts to humans. Am J Ther. 2001;8(5):345–57. doi:10.1097/00045391-200109000-00008.; https://www.gynecology.su/jour/article/view/2053Test

    الإتاحة: https://doi.org/10.17749/2313-7347/ob.gyn.rep.2024.51210.3390/nu709538810.3402/fnr.v59.2597410.1155/2018/904169410.17749/2313-7347.2017.11.1.036-04810.1152/physrev.00012.201410.3390/nu902016810.3390/nu907069710.1093/ndtplus/sfr16310.1038/ki.2012.45210.1074/jbc.M31120120010.1113/jphysiol.2004.08020010.1155/2017/679746010.1016/j.pathophys.2007.09.00510.1155/2016/260853810.1007/s00467-016-3416-310.5049/EBP.2008.6.2.8610.2147/IJNRD.S4205410.1053/ctrv.1999.009710.5049/EBP.2007.5.2.12610.2165/00002018-200528090-0000310.1016/j.maturitas.2015.08.01010.1016/0006-3223Test(94)90103-110.1177/00045632860230060710.1016/S0015-0282(98)00053-310.4236/ojog.2016.6504110.1111/j.1526-4610.1991.hed3105298.x10.1089/jwh.1998.7.115710.2165/00044011-200727010-0000410.1089/15246090031862310.17749/2313-7347.2018.12.1.054-06010.1002/14651858.CD00212410.1684/mrh.2017.041910.1080/gye.15.3.198.20110.3109/09513590.2011.57966310.1159/00005298210.1503/cmaj.12067710.1007/s00198-015-3400-y10.1007/s774-002-8445-y10.17749/2077-8333/epi.par.con.2022.11510.17749/2070-4909/farmakoekonomika.2022.15910.17749/2313-7347/ob.gyn.rep.2022.37110.17749/2077-8333/epi.par.con.2023.16810.1097/00045391-200109000-00008
    https://www.gynecology.su/jour/article/view/2053Test

  4. 4
    دورية أكاديمية

    المصدر: Obstetrics, Gynecology and Reproduction; Vol 18, No 1 (2024); 96-111 ; Акушерство, Гинекология и Репродукция; Vol 18, No 1 (2024); 96-111 ; 2500-3194 ; 2313-7347

    وصف الملف: application/pdf

    العلاقة: https://www.gynecology.su/jour/article/view/1955/1182Test; LeBleu V. Imaging the tumor microenvironment. Cancer J. 2015;21(3):174–8. https://doi.org/10.1097/PPO.0000000000000118Test.; Del Prete A., Schioppa T., Tiberio L. et al. Leukocyte trafficking in tumor microenvironment. Curr Opin Pharmacol. 2017;35:40–7. https://doi.org/10.1016/j.coph.2017.05.004Test.; Desai A., Small E.J. Treatment of advanced renal cell carcinoma patients with cabozantinib, an oral multityrosine kinase inhibitor of MET, AXL and VEGF receptors. Future Oncol. 2019;15(20):2337–48. https://doi.org/10.2217/fon-2019-0021Test.; Mantovani A., Allavena P., Sica A., Balkwill F. Cancer-related inflammation. Nature. 2008;454(7203):436–44. https://doi.org/10.1038/nature07205Test.; Torre L.A., Bray F., Siegel R.L. et al. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65(2):87–108. https://doi.org/10.3322/caac.21262Test.; Hanahan D., Coussens L.M. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21(3):309– 22. https://doi.org/10.1016/j.ccr.2012.02.022Test.; Hinshaw D.C., Shevde L.A. The tumor microenvironment innately modulates cancer progression. Cancer Res. 2019;79(18):4557–66. https://doi.org/10.1158/0008-5472.CAN-18-3962Test.; Pottier C., Wheatherspoon A., Roncarati P. et al. The importance of the tumor microenvironment in the therapeutic management of cancer. Expert Rev Anticancer Ther. 2015;15(8):943–54. https://doi.org/10.1586/14737140.2015.1059279Test.; Angell H., Galon J. From the immune contexture to the Immunoscore: the role of prognostic and predictive immune markers in cancer. Curr Opin Immunol. 2013;25(2):261–7. https://doi.org/10.1016/j.coi.2013.03.004Test.; Wang T., Niu G., Kortylewski M. et al. Regulation of the innate and adaptive immune responses by Stat-3 signaling in tumor cells. Nat Med. 2004;10(1):48–54. https://doi.org/10.1038/nm976Test.; Maimela N.R., Liu S., Zhang Y. Fates of CD8+ T cells in tumor microenvironment. Comput Struct Biotechnol J. 2019;17:1–13. https://doi.org/10.1016/j.csbj.2018.11.004Test.; Lv L., Pan K., Li X.-d. et al. The accumulation and prognosis value of tumor infiltrating IL-17 producing cells in esophageal squamous cell carcinoma. PloS One. 2011;6(3):e18219. https://doi.org/10.1371/journal.pone.0018219Test.; Plitas G., Rudensky A.Y. Regulatory T cells in cancer. Annu Rev Cancer Biol. 2020;4(1):459–77. https://doi.org/10.1146/annurevcancerbio-030419-033428Test.; Curiel T.J., Coukos G., Zou L. et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004;10(9):942–9. https://doi.org/10.1038/nm1093Test.; Fozza C., Longinotti M. T-cell traffic jam in Hodgkin's lymphoma: pathogenetic and therapeutic implications. Adv Hematol. 2011;2011:501659. https://doi.org/10.1155/2011/501659Test.; Koreishi A.F., Saenz A.J., Persky D.O. et al. The role of cytotoxic and regulatory T-cells in relapsed/refractory Hodgkin lymphoma. Appl Immunohistochem Mol Morphol. 2010;18(3):206–11. https://doi.org/10.1097/PAI.0b013e3181c7138bTest.; Gomes A.Q., Martins D.S., Silva-Santos B. Targeting γδ T lymphocytes for cancer immunotherapy: from novel mechanistic insight to clinical application. Cancer Res. 2010;70(24):10024–7. https://doi.org/10.1158/0008-5472.CAN-10-3236Test.; Tanaka M., Iwakiri Y. The hepatic lymphatic vascular system: structure, function, markers, and lymphangiogenesis. Cell Mol Gastroenterol Hepatol. 2016;2(6):733–49. https://doi.org/10.1016/j.jcmgh.2016.09.002Test.; Milne K., Köbel M., Kalloger S.E. et al. Systematic analysis of immune infiltrates in high-grade serous ovarian cancer reveals CD20, FoxP3 and TIA-1 as positive prognostic factors. PloS One. 2009;4(7):e6412. https://doi.org/10.1371/journal.pone.0006412Test.; Andreu P., Johansson M., Affara N.I. et al. FcRγ activation regulates inflammation-associated squamous carcinogenesis. Cancer Cell. 2010;17(2):121–34. https://doi.org/10.1016/j.ccr.2009.12.019Test.; Mauri C., Bosma A. Immune regulatory function of B cells. Annu Rev Immunol. 2012;30:221–41. https://doi.org/10.1146/annurevimmunol-020711-074934Test.; Horikawa M., Minard-Colin V., Matsushita T., Tedder T. F. Regulatory B cell production of IL-10 inhibits lymphoma depletion during CD20 immunotherapy in mice. J Clin Invest. 2011;121(11):4268–80. https://doi.org/10.1172/JCI59266Test.; Sharonov G.V., Serebrovskaya E.O., Yuzhakova D.V. et al. B cells, plasma cells and antibody repertoires in the tumour microenvironment. Nat Rev Immunol. 2020;20(5):294–307. https://doi.org/10.1038/s41577-019-0257-xTest.; Marcus A., Gowen B. G., Thompson T.W. et al. Recognition of tumors by the innate immune system and natural killer cells. Adv Immunol. 2014;122:91–128. https://doi.org/10.1016/B978-0-12-800267-4.00003-1Test.; Qian B.-Z., Pollard J.W. Macrophage diversity enhances tumor progression and metastasis. Cell. 2010;141(1):39–51. https://doi.org/10.1016/j.cell.2010.03.014Test.; Condeelis J., Pollard J.W. Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell. 2006;124(2):263–6. https://doi.org/10.1016/j.cell.2006.01.007Test.; Wang S.-C., Hong J.-H., Hsueh C., Chiang C.-S. Tumor-secreted SDF-1 promotes glioma invasiveness and TAM tropism toward hypoxia in a murine astrocytoma model. Lab Invest. 2012;92(1):151–62. https://doi.org/10.1038/labinvest.2011.128Test.; Franklin R.A., Liao W., Sarkar A. et al. The cellular and molecular origin of tumor-associated macrophages. Science. 2014;344(6186):921–5. https://doi.org/10.1126/science.1252510Test.; Gabrilovich D.I., Ostrand-Rosenberg S., Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol. 2012;12(4):253–68. https://doi.org/10.1038/nri3175Test.; Meredith M.M., Liu K., Darrasse-Jeze G. et al. Expression of the zinc finger transcription factor zDC (Zbtb46, Btbd4) defines the classical dendritic cell lineage. J Exp Med. 2012;209(6):1153–65. https://doi.org/10.1084/jem.20112675Test.; Nozawa H., Chiu C., Hanahan D. Infiltrating neutrophils mediate the initial angiogenic switch in a mouse model of multistage carcinogenesis. Proc Natl Acad Sci U S A. 2006;103(33):12493–8. https://doi.org/10.1073/pnas.0601807103Test.; Youn J.-I., Gabrilovich D.I. The biology of myeloid-derived suppressor cells: the blessing and the curse of morphological and functional heterogeneity. Eur J Immunol. 2010;40(11):2969–75. https://doi.org/10.1002/eji.201040895Test.; Erler J.T., Bennewith K.L., Cox T.R. et al. Hypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. Cancer Cell. 2009;15(1):35–44. https://doi.org/10.1016/j.ccr.2008.11.012Test.; Granot Z., Henke E., Comen E.A. et al. Tumor entrained neutrophils inhibit seeding in the premetastatic lung. Cancer Cell. 2011;20(3):300–14. https://doi.org/10.1016/j.ccr.2011.08.012Test.; Walker C., Mojares E., del Río Hernández A. Role of extracellular matrix in development and cancer progression. Int J Mol Sci. 2018;19(10):3028. https://doi.org/10.3390/ijms19103028Test.; Nieman K.M., Kenny H.A., Penicka C.V. et al. Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nat Med. 2011;17(11):1498–503. https://doi.org/10.1038/nm.2492Test.; Alitalo K. The lymphatic vasculature in disease. Nat Med. 2011;17(11):1371–80. https://doi.org/10.1038/nm.2545Test.; Swartz M.A., Lund A.W. Lymphatic and interstitial flow in the tumour microenvironment: linking mechanobiology with immunity. Nat Rev Cancer. 2012;12(3):210–9. https://doi.org/10.1038/nrc3186Test.; Erez N., Truitt M., Olson P. et al. Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-κB-dependent manner. Cancer Cell. 2010;17(2):135–47. https://doi.org/10.1016/j.ccr.2009.12.041Test.; Xing F., Saidou J., Watabe K. Cancer associated fibroblasts (CAFs) in tumor microenvironment. Front Biosci. 2010;15(1):166–79. https://doi.org/10.2741/3613Test.; Korneev K.V., Atretkhany K.-S. N., Drutskaya M. S. et al. TLR-signaling and proinflammatory cytokines as drivers of tumorigenesis. Cytokine. 2017;89:127–35. https://doi.org/10.1016/j.cyto.2016.01.021Test.; Shiga K., Hara M., Nagasaki T. et al. Cancer-associated fibroblasts: their characteristics and their roles in tumor growth. Cancers. 2015;7(4):2443– 58. https://doi.org/10.3390/cancers7040902Test.; Li B., Wang J. H.-C. Fibroblasts and myofibroblasts in wound healing: force generation and measurement. J Tissue Viability. 2011;20(4):108–20. https://doi.org/10.1016/j.jtv.2009.11.004Test.; Kraman M., Bambrough P.J., Arnold J.N. et al. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-α. Science. 2010;330(6005):827–30. https://doi.org/10.1126/science.1195300Test.; Armulik A., Genové G., Betsholtz C. Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell. 2011;21(2):193–215. https://doi.org/10.1016/j.devcel.2011.07.001Test.; Cooke V.G., LeBleu V.S., Keskin D.N et al. Pericyte depletion results in hypoxia-associated epithelial-to-mesenchymal transition and metastasis mediated by met signaling pathway. Cancer Cell. 2012;21(1):66–81. https://doi.org/10.1016/j.ccr.2011.11.024Test.; Turley S.J., Cremasco V., Astarita J.L. Immunological hallmarks of stromal cells in the tumour microenvironment. Nat Rev Immunol. 2015;15(11):669–82. https://doi.org/10.1038/nri3902Test.; McAndrews K.M., McGrail D.J., Ravikumar N., Dawson M.R. Mesenchymal stem cells induce directional migration of invasive breast cancer cells through TGF-β. Sci Rep. 2015;5(1):16941. https://doi.org/10.1038/srep16941Test.; Lam P.Y. Biological effects of cancer-secreted factors on human mesenchymal stem cells. Stem Cell Res Ther. 2013;4(6):138. https://doi.org/10.1186/scrt349Test.; Hu Y., Li D., Wu A. et al. TWEAK-stimulated macrophages inhibit metastasis of epithelial ovarian cancer via exosomal shuttling of microRNA. Cancer Lett. 2017;393:60–7. https://doi.org/10.1016/j.canlet.2017.02.009Test.; Farnie G., Sotgia F., Lisanti M.P. High mitochondrial mass identifies a sub-population of stem-like cancer cells that are chemo-resistant. Oncotarget. 2015;6(31):30472–86. https://doi.org/10.18632/oncotarget.5401Test.; Feig C., Jones J.O., Kraman M. et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with antiPD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci U S A. 2013;110(50):20212–7. https://doi.org/10.1073/pnas.1320318110Test.; Henke E., Nandigama R., Ergün S. Extracellular matrix in the tumor microenvironment and its impact on cancer therapy. Front Mol Biosci. 2020;6:160. https://doi.org/10.3389/fmolb.2019.00160Test.; Vaupel P., Mayer A. Hypoxia in tumors: pathogenesis-related classification, characterization of hypoxia subtypes, and associated biological and clinical implications. Adv Exp Med Biol. 2014;812:19–24. https://doi.org/10.1007/978-1-4939-0620-8_3Test.; Elinav E., Garrett W.S., Trinchieri G., Wargo J. The cancer microbiome. Nat Rev Cancer. 2019;19(7):371–6. https://doi.org/10.1038/s41568-019-0155-3Test.; Hofer H.R., Tuan R.S. Secreted trophic factors of mesenchymal stem cells support neurovascular and musculoskeletal therapies. Stem Cell Res Ther. 2016;7(1):131. https://doi.org/10.1186/s13287-016-0394-0Test.; Altman J.B., Benavides A.D., Das R., Bassiri H. Antitumor responses of invariant natural killer T cells. J Immunol Res. 2015;2015:652875. https://doi.org/10.1155/2015/652875Test.; Keely P.J. Mechanisms by which the extracellular matrix and integrin signaling act to regulate the switch between tumor suppression and tumor promotion. J Mammary Gland Biol Neoplasia. 2011;16(3):205–19. https://doi.org/10.1007/s10911-011-9226-0Test.; Guan J., Chen J. Mesenchymal stem cells in the tumor microenvironment. Biomed Rep. 2013;1(4):517–21. https://doi.org/10.3892/br.2013.103Test.; Metzler K.D., Fuchs T.A., Nauseef W.M. et al. Myeloperoxidase is required for neutrophil extracellular trap formation: implications for innate immunity. Blood. 2011;117(3):953–9. https://doi.org/10.1182/blood2010-06-290171Test.; Acuff H.B., Carter K.J., Fingleton B. et al. Matrix metalloproteinase-9 from bone marrow-derived cells contributes to survival but not growth of tumor cells in the lung microenvironment. Cancer Res. 2006;66(1):259–66. https://doi.org/10.1158/0008-5472.CAN-05-2502Test.; Pahler J.C., Tazzyman S., Erez N. et al. Plasticity in tumor-promoting inflammation: impairment of macrophage recruitment evokes a compensatory neutrophil response. Neoplasia. 2008;10(4):329–40. https://doi.org/10.1593/neo.07871Test.; Cools-Lartigue J., Spicer J., McDonald B. et al. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J Clin Invest. 2013;123(8):3446–58. https://doi.org/10.1172/JCI67484Test.; Romson J.L., Hook B., Rigot V. et al. The effect of ibuprofen on accumulation of indium-111-labeled platelets and leukocytes in experimental myocardial infarction. Circulation. 1982;66(5):1002–11. https://doi.org/10.1161/01.cir.66.5.1002Test.; Goh C.Y., Patmore S., Smolenski A. et al. The role of von Willebrand factor in breast cancer metastasis. Transl Oncol. 2021;14(4):101033. https://doi.org/10.1016/j.tranon.2021.101033Test.; Price L.C., Wort S.J. Earlier diagnosis and international registries may improve outcomes in pulmonary tumour thrombotic microangiopathy. Eur Respir J. 2016;47(2):690–1. https://doi.org/10.1183/13993003.01736-2015Test.; Farge D., Bounameaux H., Brenner B. et al. International clinical practice guidelines including guidance for direct oral anticoagulants in the treatment and prophylaxis of venous thromboembolism in patients with cancer. Lancet Oncol. 2016;17(10):e452–e466. https://doi.org/10.1016/S1470-2045Test(16)30369-2.; Tinholt M., Viken M.K., Dahm A.E. et al. Increased coagulation activity and genetic polymorphisms in the F5, F10 and EPCR genes are associated with breast cancer: a case-control study. BMC Cancer. 2014;14:845. https://doi.org/10.1186/1471-2407-14-845Test.; Pihusch R., Danzl G., Scholz M. et al. Impact of thrombophilic gene mutations on thrombosis risk in patients with gastrointestinal carcinoma. Cancer. 2002;94(12):3120–6. https://doi.org/10.1002/cncr.10590Test.; Tavares V., Pinto R., Assis J. et al. Dataset of GWAS-identified variants underlying venous thromboembolism susceptibility and linkage to cancer aggressiveness. Data Brief. 2020;30:105399. https://doi.org/10.1016/j.dib.2020.105399Test.; Vossen C.Y., Hoffmeister M., Chang-Claude J.C. et al. Clotting factor gene polymorphisms and colorectal cancer risk. J Clin Oncol. 2011;29(13):1722–7. https://doi.org/10.1200/JCO.2010.31.8873Test.; de Haas E.C., Zwart N., Meijer C. et al. Association of PAI-1 gene polymorphism with survival and chemotherapy-related vascular toxicity in testicular cancer. Cancer. 2010;116(24):5628–36. https://doi.org/10.1002/cncr.25300Test.; Duffy M.J., McGowan P.M., Harbeck N. et al. uPA and PAI-1 as biomarkers in breast cancer: validated for clinical use in level-of-evidence-1 studies. Breast Cancer Res. 2014;16(4):428. https://doi.org/10.1186/s13058-014-0428-4Test.; Tavares V., Pinto R., Assis J. et al. Venous thromboembolism GWAS reported genetic makeup and the hallmarks of cancer: Linkage to ovarian tumour behaviour. Biochim Biophys Acta Rev Cancer. 2020;1873(1):188331. https://doi.org/10.1016/j.bbcan.2019.188331Test.; Vila P., Hernandez M., Lopez-Fernandez M., Batlle J. Prevalence, follow-up and clinical significance of the anticardiolipin antibodies in normal subjects. Thromb Haemost. 1994;72(8):209–13.; Vassalo J., Spector N., de Meis E. et al. Antiphospholipid antibodies in critically ill patients with cancer: a prospective cohort study. J Crit Care. 2014;29(4):533–8. https://doi.org/10.1016/j.jcrc.2014.02.005Test.; Abdel-Wahab N., Tayar J.H., Fa'ak F. et al. Systematic review of observational studies reporting antiphospholipid antibodies in patients with solid tumors. Blood Adv. 2020;4(8):1746–55. https://doi.org/10.1182/bloodadvances.2020001557Test.; Cervera R., Rodríguez-Pintó I., Colafrancesco S. et al. 14th international congress on antiphospholipid antibodies task force report on catastrophic antiphospholipid syndrome. Autoimmun Rev. 2014;13(7):699–707. https://doi.org/10.1016/j.autrev.2014.03.002Test.; https://www.gynecology.su/jour/article/view/1955Test

  5. 5
  6. 6
    دورية أكاديمية

    المصدر: Obstetrics, Gynecology and Reproduction; Online First ; Акушерство, Гинекология и Репродукция; Online First ; 2500-3194 ; 2313-7347

    وصف الملف: application/pdf

    العلاقة: https://www.gynecology.su/jour/article/view/2088/1216Test; Thachil J., Khorana A., Carrier M. Similarities and perspectives on the two C’s – Cancer and COVID-19. J Thromb Haemost. 2021;19(5):1161–7. https://doi.org/10.1111/jth.15294Test.; Sciaudone A., Corkrey H., Humphries F., Koupenova M. Platelets and SARS-CoV-2 during COVID-19: immunity, thrombosis, and beyond. Circ Res. 2023;132(10):1272–89. https://doi.org/10.1161/CIRCRESAHA.122.321930Test.; Vassiliou A.G., Vrettou C.S., Keskinidou C. et al. Endotheliopathy in acute COVID-19 and long COVID. Int J Mol Sci. 2023;24(9):8237. https://doi.org/10.3390/ijms24098237Test.; Falanga A., Marchetti M. Cancer-associated thrombosis: enhanced awareness and pathophysiologic complexity. J Thromb Haemost. 2023;21(6):1397–408. https://doi.org/10.1016/j.jtha.2023.02.029Test.; Chandra A., Chakraborty U., Ghosh S., Dasgupta S. Anticoagulation in COVID-19: current concepts and controversies. Postgrad Med J. 2022;98(1159):395–402. https://doi.org/10.1136/postgradmedj-2021-139923Test.; Farge D., Frere C., Connors J.M. et al. International Initiative on Thrombosis and Cancer (ITAC) advisory panel. 2022 international clinical practice guidelines for the treatment and prophylaxis of venous thromboembolism in patients with cancer, including patients with COVID-19. Lancet Oncol. 2022;23(7):e334–e347. https://doi.org/10.1016/S1470-2045Test(22)00160-7.; Lecumberri R., Marcos-Jubilar M., Guillén C. Thromboprophylaxis in patients with cancer and COVID-19. Arch Bronconeumol. 2022;58(11):744–5. https://doi.org/10.1016/j.arbres.2022.08.006Test.; Gulati S., Hsu C.Y., Shah S. et al. COVID-19 and Cancer Consortium. Systemic anticancer therapy and thromboembolic outcomes in hospitalized patients with cancer and COVID-19. JAMA Oncol. 2023;9(10):1390–400. https://doi.org/10.1001/jamaoncol.2023.2934Test.; Временные методические рекомендации Министерства здравоохранения Российской Федерации. Профилактика, диагностика и лечение новой коронавирусной инфекции (COVID-19). Версия 7 (03.06.2020). М.: Министерство здравоохранения Российской Федерации, 2020, 166 с. Режим доступа: https://static-0.minzdrav.gov.ru/system/attachments/attaches/000/050/584/original/03062020_TestМR_COVID-19_v7.pdf. [Дата обращения: 15.04.2024].; Временные методические рекомендации Министерства здравоохранения Российской Федерации. Профилактика, диагностика и лечение новой коронавирусной инфекции (COVID-19). Версия 8 (03.09.2020). М.: Министерство здравоохранения Российской Федерации, 2020. 227 с. Режим доступа: https://static-0.minzdrav.gov.ru/system/attachments/attaches/000/051/777/original/030902020_COVID-19_v8.pdfTest. [Дата обращения: 15.04.2024].; Временные методические рекомендации Министерства здравоохранения Российской Федерации. Профилактика, диагностика и лечение новой коронавирусной инфекции (COVID-19). Версия 9 (26.10.2020). М.: Министерство здравоохранения Российской Федерации, 2020. 236 с. Режим доступа: https://static-0.minzdrav.gov.ru/system/attachments/attaches/000/052/550/original/MP_COVID-19_%28v9%29.pdf?1603788097Test. [Дата обращения: 15.04.2024].; Временные методические рекомендации Министерства здравоохранения Российской Федерации. Профилактика, диагностика и лечение новой коронавирусной инфекции (COVID-19). Версия 10 (08.02.2021). М.: Министерство здравоохранения Российской Федерации, 2021. 262 с. Режим доступа: https://static-0.minzdrav.gov.ru/system/attachments/attaches/000/054/662/originalTest/Временные_МР_COVID-19_%28v.10%29.pdf. [Дата обращения: 15.04.2024].; Временные методические рекомендации Министерства здравоохранения Российской Федерации. Профилактика, диагностика и лечение новой коронавирусной инфекции (COVID-19). Версия 11 (07.05.2021). М.: Министерство здравоохранения Российской Федерации, 2021. 225 с. Режим доступа: https://rmapo.ru/uploads/koronaTest/МР_COVID-19-v11.pdf. [Дата обращения: 15.04.2024].; Временные методические рекомендации Министерства здравоохранения Российской Федерации. Профилактика, диагностика и лечение новой коронавирусной инфекции (COVID-19). Версия 12 (21.09.2021). М.: Мистерство здравоохранения Российской Федерации, 2021. 232 с. Режим доступа: https://static-0.minzdrav.gov.ru/system/attachments/attaches/000/058/075/originalTest/ВМР_COVID-19_V12.pdf. [Дата обращения: 15.04.2024].; Временные методические рекомендации Министерства здравоохранения Российской Федерации. Профилактика, диагностика и лечение новой коронавирусной инфекции (COVID-19). Версия 13 (14.10.2021). М.: Министерство здравоохранения Российской Федерации, 2021. 237 с. Режим доступа: https://static-0.minzdrav.gov.ru/system/attachments/attaches/000/058/211/original/BMP-13.pdfTest. [Дата обращения: 15.04.2024].; Временные методические рекомендации Министерства здравоохранения Российской Федерации. Профилактика, диагностика и лечение новой коронавирусной инфекции (COVID-19). Версия 14 (27.12.2021). М.: Министерство здравоохранения Российской Федерации, 2021. 233 с. Режим доступа: https://static-0.minzdrav.gov.ru/system/attachments/attaches/000/059/041/originalTest/ВМР_COVID-19_V14_27-12-2021.pdf. [Дата обращения: 15.04.2024].; Временные методические рекомендации Министерства здравоохранения Российской Федерации. Профилактика, диагностика и лечение новой коронавирусной инфекции (COVID-19). Версия 15 (22.02.2022). М.: Министерство здравоохранения Российской Федерации, 2022. 245 с. Режим доступа: https://static-0.minzdrav.gov.ru/system/attachments/attaches/000/059/392/originalTest/ВМР_COVID-19_V15.pdf. [Дата обращения: 15.04.2024].; Временные методические рекомендации Министерства здравоохранения Российской Федерации. Профилактика, диагностика и лечение новой коронавирусной инфекции (COVID-19). Версия 16 (18.08.2022). М.: Министерство здравоохранения Российской Федерации, 2022. 249 с. Режим доступа: https://static-0.minzdrav.gov.ru/system/attachments/attaches/000/060/193/originalTest/ВМР_COVID-19_V16.pdf. [Дата обращения: 15.04.2024].; Léonard-Lorant I., Delabranche X., Séverac F. et al. Acute pulmonary embolism in patients with COVID-19 at CT angiography and relationship to d-Dimer levels. Radiology. 2020;296(3):E189–E191. https://doi.org/10.1148/radiol.2020201561Test.; Brito-Dellan N., Tsoukalas N., Font C. Thrombosis, cancer, and COVID-19. Support Care Cancer. 2022;30(10):8491–500. https://doi.org/10.1007/s00520-022-07098-zTest.; Yang F., Shi S., Zhu J. et al. Clinical characteristics and outcomes of cancer patients with COVID-19. J Med Virol. 2020;92(10):2067–73. https://doi.org/10.1002/jmv.25972Test.; Rüthrich M.M., Giessen-Jung C., Borgmann S. et al.; LEOSS Study Group. COVID-19 in cancer patients: clinical characteristics and outcome-an analysis of the LEOSS registry. Ann Hematol. 2021;100(2):383–93. https://doi.org/10.1007/s00277-020-04328-4Test.; Nopp S., Moik F., Jilma B. et al. Risk of venous thromboembolism in patients with COVID-19: a systematic review and meta-analysis. Res Pract Thromb Haemost. 2020;4(7):1178–91. https://doi.org/10.1002/rth2.12439Test.; Kuderer N.M., Choueiri T.K., Shah D.P. et al. COVID-19 and Cancer Consortium. Clinical impact of COVID-19 on patients with cancer (CCC19): a cohort study. Lancet. 2020;395(10241):1907–18. https://doi.org/10.1016/S0140-6736Test(20)31187-9.; Oken M.M., Creech R.H., Tormey D.C. et al. Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol. 1982;5(6):649–55.; Patell R., Bogue T., Bindal P. et al. Incidence of thrombosis and hemorrhage in hospitalized cancer patients with COVID-19. J Thromb Haemost. 2020;18(9):2349–57. https://doi.org/10.1111/jth.15018Test.; Fenioux C., Allenbach Y., Vozy A. et al. Differences of characteristics and outcomes between cancer patients and patients with no active cancer hospitalised for a SARS-CoV-2 infection. Bull Cancer. 2021;108(6):581–8. (In French). https://doi.org/10.1016/j.bulcan.2021.03.004Test.; Obispo B., Rogado J., Muñoz-Rivas N. et al.; Infanta Leonor Thrombosis Research Group. Prevalence of thrombosis in patients with cancer and SARS-CoV-2 infection. Med Clin (Barc). 2022;159(5):234–7. https://doi.org/10.1016/j.medcli.2021.08.002Test.; Li A., Kuderer N.M., Hsu C.Y. et al.; CCC19 consortium. The CoVID-TE risk assessment model for venous thromboembolism in hospitalized patients with cancer and COVID-19. J Thromb Haemost. 2021;19(10):2522–32. https://doi.org/10.1111/jth.15463Test.; https://www.gynecology.su/jour/article/view/2088Test

    الإتاحة: https://doi.org/10.17749/2313-7347/ob.gyn.rep.2024.51910.1111/jth.1529410.1161/CIRCRESAHA.122.32193010.3390/ijms2409823710.1016/j.jtha.2023.02.02910.1136/postgradmedj-2021-13992310.1016/S1470-2045Test(22)00160-710.1016/j.arbres.2022.08.00610.1001/jamaoncol.2023.293410.1148/radiol.202020156110.1007/s00520-022-07098-z10.1002/jmv.2597210.1007/s00277-020-04328-410.1002/rth2.1243910.1016/S0140-6736(20)31187-910.1111/jth.1501810.1016/j.bulcan.2021.03.00410.1016/j.medcli.2021.08.00210.1111/jth.15463
    https://www.gynecology.su/jour/article/view/2088Test

  7. 7
    دورية أكاديمية

    المساهمون: The study was not sponsored, Исследование проведено без финансовой поддержки

    المصدر: Obstetrics, Gynecology and Reproduction; Vol 18, No 2 (2024); 189-199 ; Акушерство, Гинекология и Репродукция; Vol 18, No 2 (2024); 189-199 ; 2500-3194 ; 2313-7347

    وصف الملف: application/pdf

    العلاقة: https://www.gynecology.su/jour/article/view/2050/1199Test; Brenner B. Haemostatic changes in pregnancy. Thromb Res. 2004;114(5–6):409–14. doi:10.1016/j.thromres.2004.08.004.; Cui C., Yang S., Zhang J. et al. Trimester-specific coagulation and anticoagulation reference intervals for healthy pregnancy. Thromb Res. 2017;156:82–6. doi:10.1016/j.thromres.2017.05.021.; Момот А.П., Николаева М.Г., Сердюк Г.В. и др. Оценка состояния гемостаза при физиологически протекающей беременности (методические рекомендации). Российский вестник акушера-гинеколога. 2018;18(3–2):2–37.; Pabinger I. Thrombophilia and its impact on pregnancy. Thromb Res. 2009;123 Suppl 3: S16–S21. doi:10.1016/S0049-3848(09)70128-8.; Хизроева Д.Х., Антонова А.С., Егорова Е.С., Макацария Н.А. Повторные неудачи ЭКО, тромбозы и тромбофилия. Акушерство, Гинекология и Репродукция. 2023;17(6):792–800. doi:10.17749/2313-7347/ob.gyn.rep.2023.467.; Ren K., Wei Y., Qiao R. et al. Changes in coagulation during twin pregnancies. Clin Appl Thromb Hemost. 2020;26:1076029620983898. doi:10.1177/1076029620983898.; Morikawa M., Yamada T., Turega N. et al. Coagulation-fibrinolysis is more enhanced in twin than in singleton pregnancies. J Perinat Med. 2006; 34(5):392–7. doi:10.1515/JPM.2006.078.; Lin L., Yang H., Xu Zh. et al. Explore the impact of abnormal coagulation test results on pregnancy complications and perinatal outcomes by establishing the trimester-specific reference intervals of singleton and twin pregnancies. Clin Chim Acta. 2023;541(Suppl 1):117265. doi:10.1016/j.cca.2023.117265.; Момот А.П., Молчанова И.В., Цывкина Л.П. Изменения системы гемостаза в цикле ЭКО и их влияние на эффективность процедуры. Бюллетень медицинской науки. 2017;4(8):77–81. doi:10.31684/2541-8475.2017.4(8).77-81.; Yang W., Sun Q., Zhou Z. et al. Coagulation parameters predictive of repeated implantation failure in Chinese women: a retrospective study. Medicine. 2020;99(48):e23320. doi:10.1097/MD.0000000000023320.; Westerlund E., Henriksson P., Wallén H. et al. Detection of a procoagulable state during controlled ovarian hyperstimulation for in vitro fertilization with global assays of haemostasis. Thromb Res. 2012;130(4):649–53. doi:10.1016/j.thromres.2011.11.024.; Grandone E., Di Micco P.P., Villani M. et al. Venous thromboembolism in women undergoing assisted reproductive technologies: data from the RIETE Registry. Thromb Haemost. 2018;118(11):1962–8. doi:10.1055/s-0038-1673402.; Rizwan N., Abbasi R.M., Mughal R. Maternal morbidity and perinatal outcome with twin pregnancy. J Ayub Med Coll Abbottabad. 2010;22(2):105–7.; Яковенко Е.М., Яковенко С.А. Экстракорпоральное оплодотворение и другие методы преодоления бесплодия. M., 2016. 280 c.; Liu J., Yuan E., Lee L. Gestational age-specific reference intervals for routine haemostatic assays during normal pregnancy. Clin Chim Acta. 2012;413(1–2):258–61. doi:10.1016/j.cca.2011.09.046.; Bar J., Blickstein D., Hod M. et al. Increased D-dimer levels in twin gestation. Thromb Res. 2000;98(6):485–9. doi:10.1016/s0049-3848(00)00187-0.; Yamada T., Kawaguchi S., Araki N. et al. Difference in the D-dimer rise between women with singleton and multifetal pregnancies. Thromb Res. 2013;131(6):493–6. doi:10.1016/j.thromres.2013.04.029.; Годзоева А.О., Зазерская И.Е., Власов В.С. и др. Оценка фибрин-мономера и D-димера у пациенток с бесплодием в программах вспомогательных репродуктивных технологий. Акушерство и гинекология. 2020;9:73–81. doi:10.18565/aig.2020.9.73-81.-81.; https://www.gynecology.su/jour/article/view/2050Test

    الإتاحة: https://doi.org/10.17749/2313-7347/ob.gyn.rep.2024.50710.1016/j.thromres.2004.08.00410.1016/j.thromres.2017.05.02110.1016/S0049-3848Test(09)70128-810.17749/2313-7347/ob.gyn.rep.2023.46710.1177/107602962098389810.1515/JPM.2006.07810.1016/j.cca.2023.11726510.31684/2541-8475.2017.4(8).77-8110.1097/MD.000000000002332010.1016/j.thromres.2011.11.02410.1055/s-0038-167340210.1016/j.cca.2011.09.04610.1016/s0049-3848(00)00187-010.1016/j.thromres.2013.04.02910.18565/aig.2020.9.73-81.-81
    https://www.gynecology.su/jour/article/view/2050Test

  8. 8
    دورية أكاديمية

    المصدر: Obstetrics, Gynecology and Reproduction; Vol 18, No 1 (2024); 55-67 ; Акушерство, Гинекология и Репродукция; Vol 18, No 1 (2024); 55-67 ; 2500-3194 ; 2313-7347

    وصف الملف: application/pdf

    العلاقة: https://www.gynecology.su/jour/article/view/1960/1181Test; Brinkmann V., Reichard U., Goosmann C. et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303(5663):1532–5. https://doi.org/10.1126/science.1092385Test.; Snoderly H.T., Boone B.A., Bennewitz M.F. Neutrophil extracellular traps in breast cancer and beyond: current perspectives on NET stimuli, thrombosis and metastasis, and clinical utility for diagnosis and treatment. Breast Cancer Res. 2019;21(1):145. https://doi.org/10.1186/s13058-019-1237-6Test.; Papayannopoulos V., Metzler K.D., Hakkim A., Zychlinsky A. Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps. J Cell Biol. 2010;191(3):677–91. https://doi.org/10.1083/jcb.201006052Test.; Hanahan D., Weinberg R.A. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74. https://doi.org/10.1016/j.cell.2011.02.013Test.; Balkwill F., Mantovani A. Inflammation and cancer: back to Virchow? Lancet. 2001;357(9255):539–45. https://doi.org/10.1016/S0140-6736Test(00)04046-0.; Bonavita E., Galdiero M.R., Jaillon S., Mantovani A. Phagocytes as сorrupted зolicemen in сancer-кelated шnflammation. Adv Cancer Res. 2015;128:141–71. https://doi.org/10.1016/bs.acr.2015.04.013Test.; Coffelt S., Wellenstein M., de Visser K. Neutrophils in cancer: neutral no more. Nat Rev Cancer. 2016;16(7):431–46. https://doi.org/10.1038/nrc.2016.52Test.; Colotta F., Re F., Polentarutti N. et al. Modulation of granulocyte survival and programmed cell death by cytokines and bacterial products. Blood. 1992;80(8):2012–20.; Carnevale S., Ghasemi S., Rigatelli A., Jaillon S. The complexity of neutrophils in health and disease: focus on cancer. Semin Immunol. 2020;48:101409. https://doi.org/10.1016/j.smim.2020.101409Test.; De Meo M.L., Spicer J.D. The role of neutrophil extracellular traps in cancer progression and metastasis. Semin Immunol. 2021;57:101595. https://doi.org/10.1016/j.smim.2022.101595Test.; Слуханчук Е.В., Бицадзе В.О., Солопова А.Г. и др. Маркеры внеклеточных ловушек нейтрофилов у женщин со злокачественными новообразованиями репродуктивной системы, получавших хирургическое лечение и адъювантную химиотерапию. Акушерство, Гинекология и Репродукция. 2023;17(4):420–32. https://doi.org/10.17749/2313-7347/ob.gyn.rep.2023.432Test.; Cristinziano L., Modestino L., Antonelli A. et al. Neutrophil extracellular traps in cancer. Semin Cancer Biol. 2022;79:91–104. https://doi.org/10.1016/j.semcancer.2021.07.011Test.; Sosa M.S., Bragado P., Aguirre-Ghiso J.A. Mechanisms of disseminated cancer cell dormancy: an awakening field. Nat Rev Cancer. 2014;14(9):611–22. https://doi.org/10.1038/nrc3793Test.; Albrengues J., Shields M.A., Ng D. et al. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science. 2018;361(6409):eaao4227. https://doi.org/10.1126/science.aao4227Test.; Poto R., Cristinziano L, Modestino L. et al. Neutrophil extracellular traps, angiogenesis and cancer. Biomedicines. 2022;10(2):431. https://doi.org/10.3390/biomedicines10020431Test.; Szczerba M.B., Castro-Giner F., Vetter M. et al. Neutrophils escort circulating tumour cells to enable cell cycle progression. Nature. 2019;566(7745):553–7. https://doi.org/10.1038/s41586-019-0915-yTest.; Shaul M.E., Fridlender Z.G. Tumour-associated neutrophils in patients with cancer. Nat Rev Clinical Oncol. 2019;16(10):601–20. https://doi.org/10.1038/s41571-019-0222-4Test.; Li P., Lu M., Shi J. et al. Lung mesenchymal cells elicit lipid storage in neutrophils that fuel breast cancer lung metastasis. Nat Immunol. 2020;21(11):1444–55. https://doi.org/10.1038/s41590-020-0783-5Test.; Tohme S., Yazdani H.O., Al-Khafaji A.B. et al. Neutrophil extracellular traps promote the development and progression of liver metastases after surgical stress. Cancer Res. 2016;76(6):1367–80. https://doi.org/10.1158/0008-5472.CAN-15-1591Test.; Mauracher L.M., Posch F., Martinod K. et al. Citrullinated histone H3, a biomarker of neutrophil extracellular trap formation, predicts the risk of venous thromboembolism in cancer patients. J Thromb Haemost. 2018;16(3):508–18. https://doi.org/10.1111/jth.13951Test.; Слуханчук Е.В., Бицадзе В.О., Солопова А.Г. и др. Иммунотромбоз, прогрессия опухоли и метастазирование. Роль интерлейкина-8 и внеклеточных ловушек нейтрофилов. Вопросы гинекологии, акушерства и перинатологии. 2023;22(4):48–56. https://doi.org/10.20953/1726-1678-2023-4-48-56Test.; North R.J., Neubauer R.H., Huang J.J. et al. Interleukin 1-induced, T cellmediated regression of immunogenic murine tumors. Requirement for an adequate level of already acquired host concomitant immunity. J Exp Med. 1988;168(6):2031–43. https://doi.org/10.1084/jem.168.6.2031Test.; Rébé C., Ghiringhelli F. Interleukin-1β and cancer. Cancers (Basel). 2020;12(7):1791. https://doi.org/10.3390/cancers12071791Test.; Zahorec R. Neutrophil-to-lymphocyte ratio, past, present and future perspectives. Bratisl Lek Listy. 2021;122(7):474–88. https://doi.org/10.4149/BLL_2021_078Test.; Forget P., Khalifa C., Defour J.P. et al. What is the normal value of the neutrophil-to-lymphocyte ratio? BMC Res Notes. 2017;10(1):12. https://doi.org/10.1186/s13104-016-2335-5Test.; https://www.gynecology.su/jour/article/view/1960Test

  9. 9
    دورية أكاديمية

    المصدر: Obstetrics, Gynecology and Reproduction; Vol 18, No 1 (2024); 8-22 ; Акушерство, Гинекология и Репродукция; Vol 18, No 1 (2024); 8-22 ; 2500-3194 ; 2313-7347

    وصف الملف: application/pdf

    العلاقة: https://www.gynecology.su/jour/article/view/1890/1175Test; https://www.gynecology.su/jour/article/view/1890/1176Test; Gris J.C., Mousty É., Bouvier S. et al. Increased incidence of cancer in the follow-up of obstetric antiphospholipid syndrome within the NOH-APS cohort. Haematologica. 2020;105(2):490–7. https://doi.org/10.3324/haematol.2018.213991Test.; Cabrera-Marante O., Rodríguez de Frías E., Serrano M. et al. The weight of IgA anti-β2glycoprotein I in the antiphospholipid syndrome pathogenesis: closing the gap of seronegative antiphospholipid syndrome. Int J Mol Sci. 2020;21(23):8972. https://doi.org/10.3390/ijms21238972Test.; Yoon K.H., Wong A., Shakespeare T., Sivalingam P. High prevalence of antiphospholipid antibodies in Asian cancer patients with thrombosis. Lupus. 2003;12(2):112–6. https://doi.org/10.1191/0961203303lu328oaTest.; Kansuttiviwat C., Niprapan P., Tantiworawit A. et al. Impact of antiphospholipid antibodies on thrombotic events in ambulatory cancer patients. PLoS One. 2023;18(1):e0279450. https://doi.org/10.1371/journal.pone.0279450Test.; Vassalo J., Spector N., de Meis E. et al. Antiphospholipid antibodies in critically ill patients with cancer: a prospective cohort study. J Crit Care. 2014;29(4):533–8. https://doi.org/10.1016/j.jcrc.2014.02.005Test.; Gómez-Puerta J.A., Cervera R., Espinosa G. et al. Antiphospholipid antibodies associated with malignancies: clinical and pathological characteristics of 120 patients. Semin Arthritis Rheum. 2006;35(5):322– 32. https://doi.org/10.1016/j.semarthrit.2005.07.003Test.; Sawamura M., Yamaguchi S., Murakami H. et al. Multiple autoantibody production in a patient with splenic lymphoma. Ann Hematol. 1994;68(5):251–4. https://doi.org/10.1007/BF01737426Test.; Tincani A., Taraborelli M., Cattaneo R. Antiphospholipid antibodies and malignancies. Autoimmun Rev. 2010;9(4):200–2. https://doi.org/10.1016/j.autrev.2009.04.001Test.; Benvenuto M., Mattera R., Masuelli L. et al. The crossroads between cancer immunity and autoimmunity: antibodies to self antigens. Front Biosci (Landmark Ed). 2017;22(8):1289–329. https://doi.org/10.2741/4545Test.; Cuadrado M.J., Buendía P., Velasco F. et al. Vascular endothelial growth factor expression in monocytes from patients with primary antiphospholipid syndrome. J Thromb Haemost. 2006;4(11):2461–9. https://doi.org/10.1111/j.1538-7836.2006.02193.xTest.; Wu Y.Y., Nguyen A.V., Wu X.X. et al. Antiphospholipid antibodies promote tissue factor-dependent angiogenic switch and tumor progression. Am J Pathol. 2014;184(12):3359–75. https://doi.org/10.1016/j.ajpath.2014.07.027Test.; Viall C.A., Chen Q., Liu B.et al. Antiphospholipid antibodies internalised by human syncytiotrophoblast cause aberrant cell death and the release of necrotic trophoblast debris. J Autoimmun. 2013;47:45–57. https://doi.org/10.1016/j.jaut.2013.08.005Test.; Nocella C., Bartimoccia S., Cammisotto V. et al.; SMiLe Group. Oxidative stress in the pathogenesis of antiphospholipid syndrome: implications for the atherothrombotic process. Antioxidants (Basel). 2021;10(11):1790. https://doi.org/10.3390/antiox10111790Test.; Štok U., Čučnik S., Sodin-Šemrl S., Žigon P. Extracellular vesicles and antiphospholipid syndrome: state-of-the-art and future challenges. Int J Mol Sci. 2021;22(9):4689. https://doi.org/10.3390/ijms22094689Test.; Kogure A., Yoshioka Y., Ochiya T. Extracellular vesicles in cancer metastasis: potential as therapeutic targets and materials. Int J Mol Sci. 2020;21(12):4463. https://doi.org/10.3390/ijms21124463Test.; Kalluri R., McAndrews K.M. The role of extracellular vesicles in cancer. Cell. 2023;186(8):1610–26. https://doi.org/10.1016/j.cell.2023.03.010Test.; Kasthuri R.S., Taubman M.B., Mackman N. Role of tissue factor in cancer. J Clin Oncol. 2009;27(29):4834–8. https://doi.org/10.1200Test/ JCO.2009.22.6324.; Khorana A.A., Mackman N., Falanga, A. et al. Cancer-associated venous thromboembolism. Nat Rev Dis Primers. 2022;8(1):11. https://doi.org/10.1038/s41572-022-00336-yTest.; Abu Zaanona M.I., Mantha S. Cancer-associated thrombosis. 2023 Jul 17. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing, 2023 Jan. 20. Mukai M., Oka T. Mechanism and management of cancer-associated thrombosis. J Cardiol. 2018;72(2):89–93. https://doi.org/10.1016/j.jjcc.2018.02.011Test.; Dambrauskienė R., Gerbutavičius R., Rudžianskienė M. et al. Antiphospholipid antibodies and the risk of thrombosis in myeloproliferative neoplasms. Open Life Sciences. 2023;18(1):20220545. https://doi.org/10.1515/biol-2022-0545Test; Trousseau A. Phlegmasia alba dolens Clinique Medical de L’Hotel-Dieu deParis, Vol. 3. The New Sydenham Society, London, 1865. 94 p.; Metharom P., Falasca M., Berndt M.C. The history of Armand Trousseau and cancer-associated thrombosis. Cancers (Basel). 2019;11(2):158. https://doi.org/10.3390/cancers11020158Test.; Слуханчук Е.В., Бицадзе В.О., Солопова А.Г. и др. Взаимодействие внеклеточных ловушек нейтрофилов и антифосфолипидных антител у онкологических больных. Вопросы гинекологии, акушерства и перинатологии. 2023;22(3):54–62. https://doi.org/10.20953/1726-1678-2023-3-54-62Test.; Zuckerman E., Toubi E., Golan T.D. et al. Increased thromboembolic incidence in anti-cardiolipin-positive patients with malignancy. Br J Cancer. 1995;72(2):447–51. https://doi.org/10.1038/bjc.1995.353Test.; https://www.gynecology.su/jour/article/view/1890Test

  10. 10
    دورية أكاديمية

    المصدر: Obstetrics, Gynecology and Reproduction; Vol 17, No 6 (2023); 751-768 ; Акушерство, Гинекология и Репродукция; Vol 17, No 6 (2023); 751-768 ; 2500-3194 ; 2313-7347

    وصف الملف: application/pdf

    العلاقة: https://www.gynecology.su/jour/article/view/1856/1162Test; Beiter T., Fragasso A., HartlD., Nieß A.M. Neutrophil extracellular traps: a walk on the wild side of exercise immunology. Sports Med. 2015;45(5):625–40. https://doi.org/10.1007/s40279-014-0296-1Test.; Finazzi G. The Italian Registry of antiphospholipid antibodies. Haematologica. 1997;82(1):101–5.; Saffarzadeh M., Juenemann C., Queisser M.A. et al. Neutrophil extracellular traps directly induce epithelial and endothelial cell death: a predominant role of histones. PloS One. 2012;7(2):e32366. https://doi.org/10.1371/journal.pone.0032366Test.; Fine N., Tasevski N., McCulloch C.A. et al. The neutrophil: constant defender and first responder. Front Immunol. 2020;11:571085. https://doi.org/10.3389/fimmu.2020.571085Test.; Vilen S.-T., Nyberg P., Hukkanen M. et al. Intracellular co-localization of trypsin-2 and matrix metalloprotease-9: possible proteolytic cascade of trypsin-2, MMP-9 and enterokinase in carcinoma. Exp Cell Res. 2008;314(4):914–26. https://doi.org/10.1016/j.yexcr.2007.10.025Test.; Антонеева И.И. Кислородзависимая антимикробная система нейтрофилов в динамике развития рака яичников. Казанский медицинский журнал. 2008;89(4):476–8.; Schauer C., Janko C., Munoz L.E. et al. Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat Med. 2014;20(5):511–7. https://doi.org/10.1038/nm.3547Test.; Shi X., Li B., Yuan Y. et al. The possible association between the presence of an MPO -463 G > A (rs2333227) polymorphism and cervical cancer risk. Pathol Res Pract. 2018;8(214):1142–8. https://doi.org/10.1016/j.prp.2018.05.018Test.; Falanga A., Rickles F.R. Pathophysiology of the thrombophilic state in the cancer patient. Semin Thromb Hemost. 1999;25(2):173–82. https://doi.org/10.1016/10.1055/s-2007-994919Test.; Fine N., Tasevski N., McCulloch C.A. et al. The neutrophil: constant defender and first responder. Front Immunol. 2020;11:571085. https://doi.org/10.1016/10.3389/fimmu.2020.571085Test.; Brinkmann V., Reichard U., Goosmann C. et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303(5663):1532–5. https://doi.org/10.1126/science.1092385Test.; Arazna M., Pruchniak M.P., Zycinska K., Demkow U. Neutrophil extracellular trap in human diseases. Adv Exp Med Biol. 2013;756:1–8. https://doi.org/10.1007/978-94-007-4549-0_1Test.; Yalavarthi S., Gould T.J., Rao A.N. et al. Release of neutrophil extracellular traps by neutrophils stimulated with antiphospholipid antibodies: a newly identified mechanism of thrombosis in the antiphospholipid syndrome. Arthritis Rheumatol. 2015;67(11):2990–3003. https://doi.org/10.1002/art.39247Test.; Demers M., Wagner D.D. NETosis: a new factor in tumor progression and cancer-associated thrombosis. Semin Thromb Hemost. 2014;40(3):277–83. https://doi.org/10.1055/s-0034-1370765Test.; Malcolm K.C., Worthen G.S. Lipopolysaccharide stimulates p38-dependent induction of antiviral genes in neutrophils independently of paracrine factors. J Biol Chem. 2003;278(18):15693–701. https://doi.org/10.1074/jbc.M212033200Test.; Perobelli S.M., Galvani R.G., Gonçalves-Silva T. et al. Plasticity of neutrophils reveals modulatory capacity. Braz J Med Biol Res. 2015;48(8):665–75. https://doi.org/10.1590/1414-431X20154524Test.; Yazdani H.O., Roy E., Comerci A.J. et al. Neutrophil extracellular traps drive mitochondrial homeostasis in tumors to augment growth. Cancer Res. 2019;79(21):5626–39. https://doi.org/10.1158/0008-5472.CAN-19-0800Test.; Yousefi S., Gold J., Andina N. et al. Catapult-like release of mitochondrial DNA by eosinophils contributes to antibacterial defense. Nat Med. 2008;14(9): 949–53. https://doi.org/10.1038/nm.1855Test.; Coussens L.M., Tinkle C.L., Hanahan D., Werb Z. MMP-9 supplied by bone marrow-derived cells contributes to skin carcinogenesis. Cell. 2000;103(3):481–90. https://doi.org/10.1016/s0092-8674Test(00)00139-2.; Mayadas T.N., Cullere X., Lowell C.A. The multifaceted functions of neutrophils. Annu Rev Pathol. 2014;9:181–218. https://doi.org/10.1146/annurev-pathol-020712-164023Test.; Al-Benna S., Shai Y., Jacobsen F., Steinstraesser L. Oncolytic activities of host defense peptides. Int J Mol Sci. 2011;12(11):8027–51. https://doi.org/10.3390/ijms12118027Test.; Cristinziano L., Luca Modestino L., Loffredo S. et al. Anaplastic thyroid cancer cells induce the release of mitochondrial extracellular DNA traps by viable neutrophils. J Immunol. 2020;204(5):1362–72. https://doi.org/10.4049/jimmunol.1900543Test.; Gupta A.K., Joshi M.B., Philippova M. et al. Activated endothelial cells induce neutrophil extracellular traps and are susceptible to NETosis-mediated cell death. FEBS Lett. 2010;584(14):3193–7. https://doi.org/10.1016/j.febslet.2010.06.006Test.; Hanahan D., Weinberg R.A. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74. https://doi.org/10.1016/j.cell.2011.02.013Test.; Cools-Lartigue J., Spicer J., McDonald B. et al. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J Clin Invest. 2013;123(8):3446–58. https://doi.org/10.1172/JCI67484Test.; Demers M., Krause D.S., Schatzberg D. et al. Cancers predispose neutrophils to release extracellular DNA traps that contribute to cancer-associated thrombosis. Proc Natl Acad Sci U S A. 2012;109(32):13076–81. https://doi.org/10.1073/pnas.1200419109Test.; Hoffmann J.H.O., Enk A.H. Neutrophil extracellular traps in dermatology: caught in the NET. J Dermatol Sci. 2016;84(1):3–10. https://doi.org/10.1016/j.jdermsci.2016.07.001Test.; Rosales C. Neutrophil: a cell with many roles in inflammation or several cell types? Front Physiol. 2018;9:113. https://doi.org/10.3389/fphys.2018.00113Test.; Kim J., Bae J.-S. Tumor-associated macrophages and neutrophils in tumor microenvironment. Mediators Inflamm. 2016;2016:6058147. https://doi.org/10.1155/2016/6058147Test.; Rayes R.F. Primary tumors induce neutrophil extracellular traps with targetable metastasis promoting effects. JCI Insight. 2019;5(16):e128008. https://doi.org/10.1172/jci.insight.128008Test.; Горудко И.В., Черкалина О.С., Соколов А.В. и др. Новые подходы к определению концентрации и пероксидазной активности миелопероксидазы в плазме крови человека. Биоорганическая химия. 2009;35(5):629–39.; Bromberg M.E., Capello M. Cancer and blood coagulation: molecular aspects. Cancer J Sci Am. 1999;5(3):132–8.; Loreto M.F., De Martinis M., Corsi M.P. et al. Coagulation and cancer: implications for diagnosis and management. Pathol Oncol Res. 2000;6(4):301–12. https://doi.org/10.1007/BF03187336Test.; Panagopoulos V., Leach D.A., Zinonos I. et al. Inflammatory peroxidases promote breast cancer progression in mice via regulation of the tumour microenvironment. Int J Oncol. 2017;50(4):1191–200. https://doi.org/10.3892/ijo.2017.3883Test.; Андрюков Б.Г., Сомова Л.М., Дробот Е.И., Матосова Е.В. Защитные стратегии нейтрофильных гранулоцитов от патогенных бактерий. Здоровье. Медицинская экология. Наука. 2017;(1):4–18. https://doi.org/10.5281/zenodo.345606Test.; Uribe-Querol E., Rosales C. Neutrophils in cancer: two sides of the same coin. J Immunol Res. 2015;2015:983698. https://doi.org/10.1155/2015/983698Test.; Солопова А.Г., Москвичёва В.С., Блбулян Т.А. и др. Актуальные вопросы профилактики, диагностики и лечения рака вульвы и влагалища. Акушерство, Гинекология и Репродукция. 2018;12(4):62–70. https://doi.org/10.17749/2313-7347.2018.12.4.062-070Test.; Shaul M.E., Fridlender Z.G. Cancer-related circulating and tumor-associated neutrophils – subtypes, sources and function. FEBS J. 2018;285(23):4316–42. https://doi.org/10.1111/febs.14524Test.; Merza M., Hartman H., Rahman M. et al. Neutrophil extracellular traps induce trypsin activation, Inflammation, and tissue damage in mice with severe acute pancreatitis. Gastroenterology. 2015;149(7):1920–31.e8. https://doi.org/10.1053/j.gastro.2015.08.026Test.; Metzler K.D., Fuchs T.A., Nauseef W.M. et al. Myeloperoxidase is required for neutrophil extracellular trap formation: implications for innate immunity. Blood. 2011;117(3):953–9. https://doi.org/10.1182/blood-2010-06-290171Test.; Pahler J.C., Tazzyman S., Erez N. et al. Plasticity in tumor-promoting inflammation: impairment of macrophage recruitment evokes a compensatory neutrophil response. Neoplasia. 2008;10(4):329–39. https://doi.org/10.1593/neo.07871Test.; Valadez-Cosmes P., Raftopoulou S., Mihalic Z.N. et al. Myeloperoxidase: growing importance in cancer pathogenesis and potential drug target. Pharmacol Ther. 2022;236:108052. https://doi.org/10.1016/j.pharmthera.2021.108052Test.; Cai H., Chuang C.Y., Hawkins C.L., Davies M.J. Binding of myeloperoxidase to the extracellular matrix of smooth muscle cells and subsequent matrix modification. Sci Rep. 2020;10(1):666. https://doi.org/10.1038/s41598-019-57299-6Test.; Нестерова И.В., Ковалева С.В., Фомичева Е.В. и др. Клинико-иммунологические параллели при неопластических заболеваниях органов пищеварения: клинические маркеры иммунодефицита и нарушения функционирования микробицидных и цитотоксических механизмов нейтрофильных гранулоцитов. XIV Международный конгресс по реабилитации в медицине и иммунореабилитации: тезисы докладов. Израиль, 2009. 2009;11(1):79a.; de Bont C.M., Eerden N., Boelens W.C., Pruijn G.J.M. Neutrophil proteases degrade autoepitopes of NET-associated proteins. Clin Exp Immunol. 2020;199(1):1–8. https://doi.org/10.1111/cei.13392Test.; López-Otín C., Matrisian L.M. Emerging roles of proteases in tumour suppression. Nat Rev Cancer. 2007;7(10):800–8. https://doi.org/10.1038/nrc2228Test.; Moali C., Hulmes D.J.S. Extracellular and cell surface proteases in wound healing: new players are still emerging. Eur J Dermatol. 2009;19(6):552–64. https://doi.org/10.1684/ejd.2009.0770Test.; Sanderson R.D., Bandari S.K., Vlodavsky I. Proteases and glycosidases on the surface of exosomes: newly discovered mechanisms for extracellular remodeling. Matrix Biol. 2019;75–76:160–9. https://doi.org/10.1016/j.matbio.2017.10.007Test.; Nyberg P., Ylipalosaari M., Sorsa T., Salo T. Trypsins and their role in carcinoma growth. Exp Cell Res. 2006;312(8):1219–28. https://doi.org/10.1016/j.yexcr.2005.12.024Test.; Vilen S.-T., Nyberg P., Hukkanen M. Intracellular co-localization of trypsin-2 and matrix metalloprotease-9: possible proteolytic cascade of trypsin-2, MMP-9 and enterokinase in carcinoma. Exp Cell Res. 2008;314(4):914–26. https://doi.org/10.1016/j.yexcr.2007.10.025Test.; Morimoto-Kamata R., Yui S. Insulin-like growth factor-1 signaling is responsible for cathepsin G-induced aggregation of breast cancer MCF-7 cells. Cancer Sci. 2017;108(8);1574–83. https://doi.org/10.1111/cas.13286Test.; Wilson T.J., Nannuru K.C., Futakuchi M. et al. Cathepsin G enhances mammary tumor-induced osteolysis by generating soluble receptor activator of nuclear factor-kappaB ligand. Cancer Res. 2008;68(14):5803–11. https://doi.org/10.1158/0008-5472.CAN-07-5889Test.; McLoed A.G., Sherrill T.P., Cheng D.-S. et al. Neutrophil-derived IL-1β impairs the efficacy of NF-κB inhibitors against lung cancer. Cell Rep. 2016;16(1):120–32. https://doi.org/10.1016/j.celrep.2016.05.085Test.; Clancy D.M., Sullivan G.P., Moran H.B.T. et al. Extracellular neutrophil proteases are efficient regulators of IL-1, IL-33, and IL-36 cytokine activity but poor effectors of microbial killing. Cell reports. 2018;22(11):2937–50. https://doi.org/10.1016/j.celrep.2018.02.062Test.; Acuff H.B., Carter K.J., Fingleton B. et al. Matrix metalloproteinase-9 from bone marrow-derived cells contributes to survival but not growth of tumor cells in the lung microenvironment. Cancer Res. 2006;66(1):259–66. https://doi.org/10.1158/0008-5472.CAN-05-2502Test.; Park J.-H., Rasch M.G., Qiu J. et al. Presence of insulin-like growth factor binding proteins correlates with tumor-promoting effects of matrix metalloproteinase 9 in breast cancer. Neoplasia. 2015;17(5):421–33. https://doi.org/10.1016/j.neo.2015.04.003Test.; Peng Z., Liu C., Victor A.R. et al. Tumors exploit CXCR4hiCD62Llo aged neutrophils to facilitate metastatic spread. Oncoimmunology. 2021;10(1);1870811. https://doi.org/10.1080/2162402X.2020.1870811Test.; Yang Q., Mas A., Diamond M.P., Al-Hendy A. The mechanism and function of epigenetics in uterine leiomyoma development. Reprod Sci. 2016;23(2):163–75. https://doi.org/10.1177/1933719115584449Test.; Audia J.E., Campbell R.M. Histone modifications and cancer. Cold Spring Harb Perspect Biol. 2016;8(4):a019521. https://doi.org/10.1101/cshperspect.a019521Test.; Podaza E., Sabbione F., Risnik D. et al. Neutrophils from chronic lymphocytic leukemia patients exhibit an increased capacity to release extracellular traps (NETs). Cancer Immunol Immunother. 2017;66(1):77–89. https://doi.org/10.1007/s00262-016-1921-7Test.; Nie M., Yang L., Bi X. et al. Neutrophil extracellular traps induced by IL8 promote diffuse large B-cell lymphoma progression via the TLR9 signaling. Clin Cancer Res. 2019;25(6):1867–79. https://doi.org/10.1158/1078-0432.CCR-18-1226Test.; Sun N., Li X., Wang Z. et al. A multiscale TiO2 nanorod array for ultrasensitive capture of circulating tumor cells. ACS Appl Mater Interfaces. 2016;8(20):12638–43. https://doi.org/10.1021/acsami.6b02178Test.; Mao Z., Zhang J., Shi Y. et al. CXCL5 promotes gastric cancer metastasis by inducing epithelial-mesenchymal transition and activating neutrophils. Oncogenesis. 2020;9(7):63. https://doi.org/10.1038/s41389-020-00249-zTest.; Teijeira A., Garasa S., Gato M. et al. CXCR1 and CXCR2 chemokine receptor agonists produced by tumors induce neutrophil extracellular traps that interfere with immune cytotoxicity. Immunity. 2020;52(5):856-871.e8. https://doi.org/10.1016/j.immuni.2020.03.001Test.; Tohme S., Yazdani H.O., Al-Khafaji A.B. et al. Neutrophil extracellular traps promote the development and progression of liver metastases after surgical stress. Cancer Res. 2016;76(6):1367–80. https://doi.org/10.1158/0008-5472.CAN-15-1591Test.; Fuchs T.A., Brill A., Duerschmied D. et al. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci U S A. 2010;107(36):15880–5. https://doi.org/10.1073/pnas.1005743107Test.; Chaffer C.L., Weinberg R.A. A perspective on cancer cell metastasis. Science. 2011;331(6024):1559–64. https://doi.org/10.1126/science.1203543Test.; Chen Y., Hu H., Tan S. et al. The role of neutrophil extracellular traps in cancer progression, metastasis and therapy. Exp Hematol Oncol. 2022;11(1):99. https://doi.org/10.1186/s40164-022-00345-3Test.; Farrera C., Fadeel B. Macrophage clearance of neutrophil extracellular traps is a silent process. J Immunol. 2013;191(5):2647–56. https://doi.org/10.4049/jimmunol.1300436Test.; Oklu R., Sheth R.A., Wong K.H.K. et al. Neutrophil extracellular traps are increased in cancer patients but does not associate with venous thrombosis. Cardiovasc Diagns Ther. 2017;7(Suppl 3):S140–S149. https://doi.org/10.21037/cdt.2017.08.01Test.; Li Y., Yang Y., Gan T. et al. Extracellular RNAs from lung cancer cells activate epithelial cells and induce neutrophil extracellular traps. Int J Oncol. 2019;55(1):69–80. https://doi.org/10.3892/ijo.2019.4808Test.; Klebanoff S.J. Myeloperoxidase: friend and foe. J Leukoc Biol. 2005;77(5):598–625. https://doi.org/10.1189/jlb.1204697Test.; Cools-Lartigue J., Spicer J., Najmeh S., Ferri L. Neutrophil extracellular traps in cancer progression. Cell Mol Life Sci. 2014;71(21):4179–94. https://doi.org/10.1007/s00018-014-1683-3Test.; Berger-Achituv S., Brinkmann V., Abed U.A. et al. A proposed role for neutrophil extracellular traps in cancer immunoediting. Front Immunol. 2013;4:48. https://doi.org/10.3389/fimmu.2013.00048Test.; Thalin C., Lundström S., Seignez C. et al. Citrullinated histone H3 as a novel prognostic blood marker in patients with advanced cancer. PLoS One. 2018;13(1):e0191231. https://doi.org/10.1371/journal.pone.0191231Test.; Cedervall J., Dragomir A., Saupe F. et al. Pharmacological targeting of peptidylarginine deiminase 4 prevents cancer-associated kidney injury in mice. Oncoimmunology. 2017;6(8):e1320009. https://doi.org/10.1080/2162402X.2017.1320009Test.; Hisada Y., Grover S.P., Maqsood A. et al. Neutrophils and neutrophil extracellular traps enhance venous thrombosis in mice bearing human pancreatic tumors. Haematologica. 2020;105(1):218–25. https://doi.org/10.3324/haematol.2019.217083Test.; Zhu T., Zou X., Yang C. et al. Neutrophil extracellular traps promote gastric cancer metastasis by inducing epithelial-mesenchymal transition. Int J Mol Med. 2021;48(1):127. https://doi.org/10.3892/ijmm.2021.4960Test.; Volkov D.V., Tetz G.V., Rubtsov Y.P. et al. Neutrophil extracellular traps (NETs): opportunities for targeted therapy. Acta Naturae. 2021;13(3):15–23. https://doi.org/10.32607/actanaturae.11503Test.; Zhang Y., Chandra V., Sanchez E.R. et al. Interleukin-17-induced neutrophil extracellular traps mediate resistance to checkpoint blockade in pancreatic cancer. J Exp Med. 2020:217(12):e20190354. https://doi.org/10.1084/jem.20190354Test.; Schalper K.A., Carleton M., Zhou M. et al. Elevated serum interleukin-8 is associated with enhanced intratumor neutrophils and reduced clinical benefit of immune-checkpoint inhibitors. Nat Med. 2020;26(5):688–92. https://doi.org/10.1038/s41591-020-0856-xTest.; https://www.gynecology.su/jour/article/view/1856Test