-
1Academic Journal
المؤلفون: A. A. Polikarpova, S. V. Ignatiev, A. V. Lyanguzov, P. A. Zharkov, O. M. Tselousova, А. А. Поликарпова, С. В. Игнатьев, А. В. Лянгузов, П. А. Жарков, О. М. Целоусова
المساهمون: The study was performed without external funding, Исследование проведено без спонсорской поддержки
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 11, № 3 (2024); 66-70 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 11, № 3 (2024); 66-70 ; 2413-5496 ; 2311-1267
مصطلحات موضوعية: оперативное лечение, factor XII deficiency, bleeding, thrombosis, surgical treatment, дефицит фактора XII, кровотечения, тромбоз
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/1073/934; Schmaier A.H., Stavrou E.X. Factor XII – What’s important but not commonly thought about. Research and Practice in Thrombosis and Haemostasis. 2019;3(4):599–606. doi:10.1002/rth2.12235.; Colman R.W. Factor XII deficiency. NORD Guide to Rare Disorders. Philadelphia, PA: Lippincott Williams and Wilkins, 2003. 895p.; Флоринский Д.Б., Левин П.А., Пшонкин A.В., Федорова Д.В., Калинина М.П., Полетаев А.В., Серегина Е.А., Грачев Н.С., Жарков П.А. Дефицит XII фактора у детей: коагулопатия или лабораторная находка? Педиатрия. Журнал им. Г.Н. Сперанского. 2022;101(2):50–5. doi:10.24110/0031-403X-2022-101-2-50-55.; Kumar R., Gupta A., Mathur P., Garg M. Factor XII deficiency ’ A rare coagulation disorder. Indian Journal of Child Health. 2016;3:349–50. doi:10.32677/IJCH.2016.v03.i04.020.; Кабаева Е.Н. Дефицит фактора свертывания FXII: риски кровотечения или тромбоза? Медвестник – информационный портал медработников Беларуси. [Электронный ресурс] URL: https://medvestnik.by/konspektvracha/defitsit-faktora-svertyvaniya-fxiiriski-krovotecheniya-ili-tromboza (дата обращения: 24.07.2024).; Stavrou E.X., Fang C., Bane K.L., Long A.T., Naudin C., Kucukal E., Gandhi A., Brett-Morris A., Mumaw M.M., Izadmehr S., Merkulova A., Reynolds C.C., Alhalabi O., Nayak L., Yu W.M., Qu C.K., Meyerson H.J., Dubyak G.R., Gurkan U.A., Nieman M.T., Sen Gupta A., Renné T., Schmaier A.H. Factor XII and uPAR upregulate neutrophil functions to influence wound healing. J Clin Invest. 2018;128(3):944–59. doi:10.1172/JCI92880.; Stavrou E., Schmaier A.H. Factor XII: what does it contribute to our understanding of the physiology and pathophysiology of hemostasis & thrombosis. Thrombosis Research. 2010;125(3):210–5. doi:10.1016/j.thromres.2009.11.028.; Konrath S., Mailer R.K., Renné T. Mechanism, Functions, and Diagnostic Relevance of FXII Activation by Foreign Surfaces. Hamostaseologie. 2021;41(6):489–501. doi:10.1055/a-1528-0499.; Long A.T., Kenne E., Jung R., Fuchs T.A., Renné T. Contact system revisited: an interface between inflammation, coagulation, and innate immunity. J Thromb Haemost. 2016;14(3):427–37. doi:10.1111/jth.13235.; Зубаиров Д.М. Молекулярные основы свертывания крови и тромбообразования. Казань: Фэн, 2000. 364 c.; Тимербаев В.Н., Киселёв С.В. Развитие представлений об инициировании свертывания крови от А.А. Шмидта до Д.М. Зубаирова. Казанский медицинский журнал. 2013;94(5):755–60.; Воробьева Н.А., Елизаров М.В., Аверьянова А.В., Путанов М.А., Шемякина Н.Я. Периоперационное ведение пациента с наследственным дефицитом фактора свертывания крови XII при кардиохирургической операции (клинический случай). Анестезиология и реаниматология. 2019;(3):97–104. doi:10.17116/anaesthesiology201903197.; Kessira A., Amireche A., Brouk H. A Rare Case of Factor XII Deficiency Manifesting as bleeding disorder. ISTH Congress Abstracts, 2022. VPB0721.; Ахмедова В.А., Шадевский В.М., Судакова А.Н., Гаус О.В., Гудалов С.О., Шустов А.В. Редкий клинический случай болезни Хагемана у пациентки кардиологического отделения. Русский медицинский журнал. 2018;6(1):46–8.; Wu X., Ding Q., Wang X., Dai J., Wu W. The prevalence of heterozygous F12 mutations in Chinese population and its relevance to incidents of thrombosis. BMC Med Genet. 2018;19(1):50. doi:10.1186/s12881-018-0557-1.; Яковлева Е.В., Демидова Е.Ю., Саломашкина В.В. Селиванова Д.С., Сурин В.Л., Горгидзе Л.А., Зозуля Н.И. Клинические проявления наследственного дефицита фактора свертывания крови XII. Гематология и трансфузиология. 2022;67(S2):89.; Al-Ansari R.Y., Alruwaili A.F., Alqahtani K.M., Al-Harbi A.F., Woodman A. Familial Multiple Coagulation Factor Deficiencies of FXI and FXII in an Asymptomatic Saudi Woman. J Invest Med High Impact Case Rep. 2023;11:23247096231199413. doi:10.1177/23247096231199413.; Fernandes H.D., Newton S., Rodrigues J.M. Factor XII Deficiency Mimicking Bleeding Diathesis: A Unique Presentation and Diagnostic Pitfall. Cureus. 2018;10(6):e2817. doi:10.7759/cureus.2817.; Работинский С.Е., Буланова Е.Л., Осокин И.П., Сизова И.Ю., Дзюба С.В. Дефицит фактора XII (фактора Хагемана) в акушерской практике: анализ двух случаев. Тромбоз, гемостаз и реология. 2022;(1):80–4. doi:10.25555/THR.2022.1.1013.; Lämmle B., Wuillemin W.A., Huber I., Krauskopf M., Zürcher C., Pflugshaupt R., Furlan M. Thromboembolism and bleeding tendency in congenital factor XII deficiency – a study on 74 subjects from 14 Swiss families. Thromb Haemost. 1991;65(2):117–21. PMID: 1905067.; Cheng Q., Tucker E.I., Pine M.S., Sisler I., Matafonov A., Sun M.F., White-Adams T.C., Smith S.A., Hanson S.R., McCarty O.J., Renné T., Gruber A., Gailani D. A role for factor XIIa-mediated factor XI activation in thrombus formation in vivo. Blood. 2010;116(19):3981–9. doi:10.1182/blood-2010-02-270918.; Matafonov A., Leung P.Y., Gailani A.E., Grach S.L., Puy C., Cheng Q., Sun M.F., McCarty O.J., Tucker E.I., Kataoka H., Renné T., Morrissey J.H., Gruber A., Gailani D. Factor XII inhibition reduces thrombus formation in a primate thrombosis model. Blood. 2014;123(11):1739–46. doi:10.1182/blood-2013-04-499111.; Tashchilova A., Podoplelova N., Sulimov A., Kutov D., Ilin I., Panteleev M., Shikhaliev K., Medvedeva S., Novichikhina N., Potapov A., Sulimov V. New Blood Coagulation Factor XIIa Inhibitors: Molecular Modeling, Synthesis, and Experimental Confirmation. Molecules. 2022;27(4):1234. doi:10.3390/molecules27041234.; Wilbs J., Kong X-D., Middendorp S.J., Prince R., Cooke A., Demarest C.T., Abdelhafez M.M., Roberts K., Umei N., Gonschorek P., Lamers C., Deyle K., Rieben R., Cook K.E., Angelillo-Scherrer A., Heinis C. Cyclic peptide FXII inhibitor provides safe anticoagulation in a thrombosis model and in artificial lungs. Nat Commun. 2020;11(1):3890. doi:10.1038/s41467-020-17648-w.; https://journal.nodgo.org/jour/article/view/1073
-
2Academic Journal
المؤلفون: T. Yu. Yafoshkina, D. B. Florinskiy, A. E. Rudneva, P. A. Levin, P. A. Zharkov, Т. Ю. Яфошкина, Д. Б. Флоринский, А. Е. Руднева, П. А. Левин, П. А. Жарков
المساهمون: The study was performed without external funding., Исследование проведено без спонсорской поддержки.
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 11, № 2 (2024); 21-27 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 11, № 2 (2024); 21-27 ; 2413-5496 ; 2311-1267
مصطلحات موضوعية: асимптоматический тромбоз у детей, post-thrombotic syndrome in children, post-thrombotic syndrome following asymptomatic thrombosis, long-term complications of deep vein thrombosis, incidental thrombosis in child, посттромботический синдром у детей, посттромботический синдром после асимптоматических тромбозов, осложнения тромбоза глубоких вен
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/1036/925; O’Brien S.H., Stanek J.R., Witmer C.M., Raffi ni L. The Continued Rise of Venous Thromboembolism Across US Children’s Hospitals. Pediatrics. 2022;149(3):e2021054649. doi:10.1542/peds.2021-054649.; Sol J.J., Knoester H., de Neef M., Smets A.M.J.B., Betlem A., van Ommen C.H. Chronic Complications After Femoral Central Venous Catheter-related Thrombosis in Critically Ill Children. J Pediatr Hematol Oncol. 2015;37(6):462–7. doi:10.1097/MPH.0000000000000370.; https://www.uptodate.com/contents/venous-thrombosis-andthromboembolism-vte-in-children-risk-factors-clinicalmanifestations-and-diagnosis.; Monagle P., Chan A.K.C., Goldenberg N.A., Ichord R.N., Journeycake J.M., Nowak-Göttl U., Vesely S.K. Antithrombotic therapy in neonates and children: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest. 2012;141(2 Suppl):e737S–e801S. doi:10.1378/chest.11-2308. Erratum in: Chest. 2014;146(6):1694. Dosage error in article text. Erratum in: Chest. 2014;146(5):1422. PMID: 22315277.; Monagle P., Cuello C.A., Augustine C., Bonduel M., Brandão L.R., Capman T., Chan A.K.C., Hanson S., Male C., Meerpohl J., Newall F., O’Brien S.H., Raffi ni L., van Ommen H., Wiernikowski J., Williams S., Bhatt M., Riva J.J., Roldan Y., Schwab N., Mustafa R.A., Vesely S.K. American Society of Hematology 2018 Guidelines for management of venous thromboembolism: treatment of pediatric venous thromboembolism. Blood Adv. 2018;2(22):3292–3316. doi:10.1182/bloodadvances.2018024786.; Жарков П.A. Антитромботическая терапия и профилактика тромбозов глубоких вен у детей с гемобластозами и синдромами костномозговой недостаточности. Автореф. дис. … д-ра мед. наук. М., 2019. 52 с.; Jones S., Monagle P., Newall F. Do asymptomatic clots in children matter? Thromb Res. 2020;189:24–34. doi:10.1016/j.thromres.2020.02.013.; Яфошкина Т.Ю., Жарков П.А. Посттромботический синдром у детей (обзор литературы). Российский журнал детской гематологии и онкологии (РЖДГиО). 2022;9(1):45–51. doi:10.21682/2311-1267-2022-9-1-45-51.; Manco-Johnson M.J., Knapp-Clevenger R., Miller B.I., Hays T. Post-thrombotic syndrome (PTS) in children: validation of a new pediatric outcome instrument and results in a comprehensive cohort of children with extremity deep vein thrombosis (DVT). Blood. 2003;102:553A–553A abstract. ISI.; Kuhle S., Koloshuk B., Marzinotto V., Bauman M., Massicotte P., Andrew M., Chan A., Abdolell M., Mitchell L. A cross-sectional study evaluating post-thrombotic syndrome in children. Thromb Res. 2003;111(4–5):227–33. doi:10.1016/j.thromres.2003.09.008. PMID: 14693168.; Avila M.L., Feldman B.M., Pullenayegum E., Lumia C., Montoya M.I., Vincelli J. Post-thrombotic syndrome in children: Measurement properties of CAPTSure, a new diagnostic tool. Res Pract Thromb Haemost. 2019;3(4):652–7. doi:10.1002/rth2.12251. PMID: 31624784.; Avila L., Amiri N., De R., Vincelli J., Pullenayegum E., Williams S., Brandão L.R. Characteristics of upper- and lower-extremity deep vein thrombosis and predictors of postthrombotic syndrome in children. Blood Adv. 2021;5(19):3737–47. doi:10.1182/bloodadvances.2021004727.; Клинические рекомендации «Острый лимфобластный лейкоз». М., 2020.; Kuhle S., Spavor M., Massicotte P., Halton J., Cherrick I., Dix D., Mahoney D., Bauman M., Desai S., Mitchell L.G. Prevalence of postthrombotic syndrome following asymptomatic thrombosis in survivors of acute lymphoblastic leukemia. J Thromb Haemost. 2008;6(4):589–94. doi:10.1111/j.1538-7836.2008.02901.x.; Avila M.L., Duan L., Cipolla A., Kim A., Kahr W.H.A., Williams S., Brandão L.R. Postthrombotic syndrome following upper extremity deep vein thrombosis in children. Blood. 2014;124(7):1166–73. doi:10.1182/blood-2014-04-570531.; Engel E.R., Nguyen A.T.H., Amankwah E.K., Albisetti M., Brandão L.R., Goldenberg N.A., Betensky M. Predictors of postthrombotic syndrome in pediatric thrombosis: A systematic review and meta-analysis of the literature. J Thromb Haemost. 2020;18(10):2601–12. doi:10.1111/jth.14984.; Жарков П.А., Шифрин Ю.А., Новичкова Г.А. Эффективность антитромботической терапии тромбозов глубоких вен у детей с заболеваниями крови. Вопросы гематологии/онкологии и иммунопатологии в педиатрии. 2019;18(1):34–42. doi:10.24287/1726-1708-2019-18-1-34-42.; Kumar R., Rodriguez V., Matsumoto J.M., Khan S.P., Weaver A.L., McBane R.D. Health-related quality of life in children and young adults with post-thrombotic syndrome: results from a cross-sectional study. Pediatr Blood Cancer. 2014;61(3):546–51. doi:10.1002/pbc.24840. PMID: 24347376.; Avila L., Betensky M., Cohen C., Ahuja S., Goldenberg N., Zia A. Clinical care of pediatric patients with or at risk of postthrombotic syndrome: guidance from the ISTH SSC Subcommittee on pediatric and neonatal thrombosis and hemostasis. J Thromb Haemost. 2024;22(2):365–78. doi:10.1016/j.jtha.2023.10.012.; https://journal.nodgo.org/jour/article/view/1036
-
3Academic Journal
المؤلفون: P. V. Kralichkin, D. Yu. Kachanov, A. V. Pshonkin, P. A. Zharkov, П. В. Краличкин, Д. Ю. Качанов, А. В. Пшонкин, П. А. Жарков
المساهمون: The study was performed without external funding., Исследование проведено без спонсорской поддержки.
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 11, № 2 (2024); 61-66 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 11, № 2 (2024); 61-66 ; 2413-5496 ; 2311-1267
مصطلحات موضوعية: ангиогенез, hemostasis, metastasis, proliferation, angiogenesis, гемостаз, метастазирование, пролиферация
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/1040/909; Lefrancais E., Ortiz-Munoz G., Caudrillier A., Mallavia B., Liu F., Sayah D.M. The lung is a site of platelet biogenesis and a reservoir for haematopoietic progenitors. Nature. 2017;544(7648):105–9. doi:10.1038/nature21706.; Holinstat M. Normal platelet function. Cancer Metastasis Rev. 2017;36:195–8. doi:10.1007/s10555-017-9677-x.; Holinstat M., Tourdot B.E. Coronary heart disease risk factors take a disproportional toll on women. Arterioscler Thromb Vasc Biol. 2015;35(4):750–1. doi:10.1161/ATVBAHA.115.305466.; Koupenova M., Mick E., Mikhalev E., Benjamin E.J., Tanriverdi K., Freedman J.E. Sex diff erences in platelet toll-like receptors and their association with cardiovascular risk factors. Arterioscler Thromb Vasc Biol. 2015;35(4):1030–7. doi:10.1161/ATVBAHA.114.304954.; Clemetson K.J. Platelets and pathogens. Cell Mol Life Sci. 2010;67(4):495–8. doi:10.1007/s00018-009-0204-2.; Stark R.J., Aghakasiri N., Rumbaut R.E. Platelet-derived Toll-like receptor 4 (Tlr-4) is suffi cient to promote microvascular thrombosis in endotoxemia. PLoS One. 2012;7(7):e41254. doi:10.1371/journal.pone.0041254.; Cox D., Kerrigan S.W., Watson S.P. Platelets and the innate immune system: mechanisms of bacterial-induced platelet activation. J Thromb Haemost. 2011;9(6):1097–107. doi:10.1111/j.1538-7836.2011.04264.x.; Elgueta R., Benson M.J., de Vries V.C., Wasiuk A., Guo Y., Noelle R.J. Molecular mechanism and function of CD40/CD40L engagement in the immune system. Immunol Rev. 2009;229(1):152–72. doi:10.1111/j.1600-065X.2009.00782.x.; Danese S., Katz J.A., Saibeni S., Papa A., Gasbarrini A., Vecchi M., Fiocchi C. Activated platelets are the source of elevated levels of soluble CD40 ligand in the circulation of infl ammatory bowel disease patients. Gut. 2003;52(10):1435–41. doi:10.1136/gut.52.10.1435.; Yacoub D., Hachem A., Théorêt J.F., Gillis M.A., Mourad W., Merhi Y. Enhanced levels of soluble CD40 ligand exacerbate platelet aggregation and thrombus formation through a CD40-dependent tumor necrosis factor receptor-associated factor-2/Rac1/p38 mitogen-activated protein kinase signaling pathway. Arterioscler Thromb Vasc Biol. 2010;30(12):2424–33. doi:10.1161/ATVBAHA.110.216143.; Semple J.W., Italiano J.E. Jr, Freedman J. Platelets and the immune continuum. Nat Rev Immunol. 2011;11(4):264–74. doi:10.1038/nri2956.; Youssefi an T., Drouin A., Massé J.M., Guichard J., Cramer E.M. Host defense role of platelets: engulfment of HIV and Staphylococcus aureus occurs in a specifi c subcellular compartment and is enhanced by platelet activation. Blood. 2002;99(11):4021–9. doi:10.1182/blood-2001-12-0191.; Maouia A., Rebetz J., Kapur R., Semple J.W. The Immune Nature of Platelets Revisited. Transfus Med Rev. 2020;34(4):209–20. doi:10.1016/j.tmrv.2020.09.005.; Wong C.H., Jenne C.N., Petri B., Chrobok N.L., Kubes P. Nucleation of platelets with blood-borne pathogens on Kupff er cells precedes other innate immunity and contributes to bacterial clearance. Nat Immunol. 2013;14(8):785–92. doi:10.1038/ni.2631.; Mishan M.A., Ahmadiankia N., Bahrami A.R. CXCR4 and CCR7: Two eligible targets in targeted cancer therapy. Cell Biol Int. 2016;40(9):955–67. doi:10.1002/cbin.10631.; Seyfried T.N., Huysentruyt L.C. On the origin of cancer metastasis. Crit Rev Oncog. 2013;18(1–2):43–73. doi:10.1615/critrevoncog.v18.i1-2.40.; Gay L.J., Felding-Habermann B. Contribution of platelets to tumour metastasis. Nat Rev Cancer. 2011;11(2):123–34. doi:10.1038/nrc3004.; Elaskalani O., Berndt M.C., Falasca M., Metharom P. Targeting Platelets for the Treatment of Cancer. Cancers (Basel). 2017;9(7):94. doi:10.3390/cancers9070094.; Melki I., Tessandier N., Zuff erey A., Boilard E. Platelet microvesicles in health and disease. Platelets. 2017;28(3):214–21. doi:10.1080/09537104.2016.1265924.; Naderi-Meshkin H., Ahmadiankia N. Cancer metastasis versus stem cell homing: Role of platelets. J Cell Physiol. 2018;233(12):9167–78. doi:10.1002/jcp.26937.; Reneman R.S., Hoeks A.P. Wall shear stress as measured in vivo: consequences for the design of the arterial system. Med Biol Eng Comput. 2008;46(5):499–507. doi:10.1007/s11517-008-0330-2.; Erpenbeck L., Schön M.P. Deadly allies: the fatal interplay between platelets and metastasizing cancer cells. Blood. 2010;115(17):3427–36. doi:10.1182/blood-2009-10-247296.; Gay L.J., Felding-Habermann B. Platelets alter tumor cell attributes to propel metastasis: programming in transit. Cancer Cell. 2011;20(5):553–4. doi:10.1016/j.ccr.2011.11.001.; Kitamura T., Qian B.Z., Pollard J.W. Immune cell promotion of metastasis. Nat Rev Immunol. 2015;15(2):73–86. doi:10.1038/nri3789.; Palumbo J.S., Degen J.L. Mechanisms linking tumor cell-associated procoagulant function to tumor metastasis. Thromb Res. 2007;120 Suppl 2: S22–8. doi:10.1016/S0049-3848(07)70127-5.; Borsig L. The role of platelet activation in tumor metastasis. Expert Rev Anticancer Ther. 2008;8(8):1247–55. doi:10.1586/14737140.8.8.1247.; Placke T., Örgel M., Schaller M., Jung G., Rammensee H.G., Kopp H.G., Salih H.R. Platelet-derived MHC class I confers a pseudonormal phenotype to cancer cells that subverts the antitumor reactivity of natural killer immune cells. Cancer Res. 2012;72(2):440–8. doi:10.1158/0008-5472.CAN-11-1872.; Gabrilovich D., Ishida T., Oyama T., Ran S., Kravtsov V., Nadaf S., Carbone D.P. Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically aff ects the diff erentiation of multiple hematopoietic lineages in vivo. Blood. 1998;92(11):4150–66. PMID: 9834220.; Laxmanan S., Robertson S.W., Wang E., Lau J.S., Briscoe D.M., Mukhopadhyay D. Vascular endothelial growth factor impairs the functional ability of dendritic cells through Id pathways. Biochem Biophys Res Commun. 2005;334(1):193–8. doi:10.1016/j.bbrc.2005.06.065.; Reddig P.J., Juliano R.L. Clinging to life: cell to matrix adhesion and cell survival. Cancer Metastasis Rev. 2005;24(3):425–39. doi:10.1007/s10555-005-5134-3.; Alfano D., Iaccarino I., Stoppelli M.P. Urokinase signaling through its receptor protects against anoikis by increasing BCL-xL expression levels. J Biol Chem. 2006;281(26):17758–67. doi:10.1074/jbc.M601812200.; Douma S., Van Laar T., Zevenhoven J., Meuwissen R., Van Garderen E., Peeper D.S. Suppression of anoikis and induction of metastasis by the neurotrophic receptor TrkB. Nature. 2004;430(7003):1034–9. doi:10.1038/nature02765.; Golebiewska E.M., Poole A.W. Platelet secretion: From haemostasis to wound healing and beyond. Blood Rev. 2015;29(3):153–62. doi:10.1016/j.blre.2014.10.003.; Sharma S.V., Bell D.W., Settleman J., Haber D.A. Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer. 2007;7(3):169–81. doi:10.1038/nrc2088.; Tanaka K., Okugawa Y., Toiyama Y., Inoue Y., Saigusa S., Kawamura M., Araki T., Uchida K., Mohri Y., Kusunoki M. Brainderived neurotrophic factor (BDNF)-induced tropomyosin-related kinase B (Trk B) signaling is a potential therapeutic target for peritoneal carcinomatosis arising from colorectal cancer. PLoS One. 2014;9(5):e96410. doi:10.1371/journal.pone.0096410.; Xiao Y.C., Yang Z.B., Cheng X.S., Fang X.B., Shen T., Xia C.F., Liu P., Qian H.H., Sun B., Yin Z.F., Li Y.F. CXCL8, overexpressed in colorectal cancer, enhances the resistance of colorectal cancer cells to anoikis. Cancer Lett. 2015;361(1):22–32. doi:10.1016/j.canlet.2015.02.021.; Zeng Q., McCauley L.K., Wang C.Y. Hepatocyte growth factor inhibits anoikis by induction of activator protein 1-dependent cyclooxygenase-2. Implication in head and neck squamous cell carcinoma progression. J Biol Chem. 2002;277(51):50137–42. doi:10.1074/jbc.M208952200.; Luey B.C., May F.E. Insulin-like growth factors are essential to prevent anoikis in oestrogen-responsive breast cancer cells: importance of the type I IGF receptor and PI3-kinase/Akt pathway. Mol Cancer. 2016;15:8. doi:10.1186/s12943-015-0482-2.; Freedman J.E., Loscalzo J., Barnard M.R., Alpert C., Keaney J.F., Michelson A.D. Nitric oxide released from activated platelets inhibits platelet recruitment. J Clin Invest. 1997;100(2):350–6. doi:10.1172/JCI119540.; Chanvorachote P., Pongrakhananon V., Chunhacha P. Prolonged nitric oxide exposure enhances anoikis resistance and migration through epithelial-mesenchymal transition and caveolin-1 upregulation. Biomed Res Int. 2014;2014:941359. doi:10.1155/2014/941359.; Bao W., Qiu H., Yang T., Luo X., Zhang H., Wan X. Upregulation of TrkB promotes epithelial-mesenchymal transition and anoikis resistance in endometrial carcinoma. PLoS One. 2013;8(7):e70616. doi:10.1371/journal.pone.0070616.; Jie X.X., Zhang X.Y., Xu C.J. Epithelial-to-mesenchymal transition, circulating tumor cells and cancer metastasis: Mechanisms and clinical applications. Oncotarget. 2017;8(46):81558–71. doi:10.18632/oncotarget.18277.; Moustakas A., Heldin C.H. Signaling networks guiding epithelialmesenchymal transitions during embryogenesis and cancer progression. Cancer Sci. 2007;98(10):1512–20. doi:10.1111/j.1349-7006.2007.00550.x.; Palena C., Hamilton D.H., Fernando R.I. Infl uence of IL-8 on the epithelial-mesenchymal transition and the tumor microenvironment. Future Oncol. 2012;8(6):713–22. doi:10.2217/fon.12.59.; Li N. Platelets in cancer metastasis: To help the “villain” to do evil. Int J Cancer. 2016;138(9):2078–87. doi:10.1002/ijc.29847.; McCarty O.J., Mousa S.A., Bray P.F., Konstantopoulos K. Immobilized platelets support human colon carcinoma cell tethering, rolling, and fi rm adhesion under dynamic fl ow conditions. Blood. 2000;96(5):1789–97. PMID: 10961878.; Janowska-Wieczorek A., Wysoczynski M., Kijowski J., MarquezCurtis L., Machalinski B., Ratajczak J., Ratajczak M.Z. Microvesicles derived from activated platelets induce metastasis and angiogenesis in lung cancer. Int J Cancer. 2005;113(5):752–60. doi:10.1002/ijc.20657.; Коваленко Т.А., Пантелеев М.А., Свешникова А.Н. Роль тканевого фактора в метастазировании, неоангиогенезе и гемостазе при онкологических заболеваниях. Онкогематология. 2019;14(2):70–85. doi:10.17650/1818-8346-2019-14-2-70-85.; Бутылин А.А., Пантелеев М.А., Атауллаханов Ф.И. Пространственная динамика свертывания крови. Российский химический журнал. 2007;51(1):45–50.; Подоплелова Н.А., Сулимов В.Б., Тащилова А.С., Ильин И.С., Пантелеев М.А., Ледeнева И.В., Шихалиев Х.С. Свертывание крови в XXI веке: новые знания, методы и перспективы для терапии. Вопросы гематологии/онкологии и иммунопатологии в педиатрии. 2020;19(1):139–57. doi:10.24287/1726-1708-2020-19-1-139-157.; Fischer E.G., Riewald M., Huang H.Y., Miyagi Y., Kubota Y., Mueller B.M., Ruf W. Tumor cell adhesion and migration supported by interaction of a receptor-protease complex with its inhibitor. J Clin Invest. 1999;104(9):1213–21. doi:10.1172/JCI7750.; Orellana R., Kato S., Erices R., Bravo M.L., Gonzalez P., Oliva B., Cubillos S., Valdivia A., Ibañez C., Brañes J., Barriga M.I., Bravo E., Alonso C., Bustamente E., Castellon E., Hidalgo P., Trigo C., Panes O., Pereira J., Mezzano D., Cuello M.A., Owen G.I. Platelets enhance tissue factor protein and metastasis initiating cell markers, and act as chemoattractants increasing the migration of ovarian cancer cells. BMC Cancer. 2015;15:290. doi:10.1186/s12885-015-1304-z.; Läubli H., Spanaus K.S., Borsig L. Selectin-mediated activation of endothelial cells induces expression of CCL5 and promotes metastasis through recruitment of monocytes. Blood. 2009;114(20):4583–91. doi:10.1182/blood-2008-10-186585.; Labelle M., Begum S., Hynes R.O. Direct signaling between platelets and cancer cells induces an epithelial-mesenchymal-like transition and promotes metastasis. Cancer Cell. 2011;20(5):576–90. doi:10.1016/j.ccr.2011.09.009.; Padua D., Zhang X.H., Wang Q., Nadal C., Gerald W.L., Gomis R.R., Massagué J. TGFbeta primes breast tumors for lung metastasis seeding through angiopoietin-like 4. Cell. 2008;133(1):66–77. doi:10.1016/j.cell.2008.01.046.; Bielenberg D.R., Zetter B.R. The Contribution of Angiogenesis to the Process of Metastasis. Cancer J. 2015;21(4):267–73. doi:10.1097/PPO.0000000000000138.; Wojtukiewicz M.Z., Sierko E., Hempel D., Tucker S.C., Honn K.V. Platelets and cancer angiogenesis nexus. Cancer Metastasis Rev. 2017;36(2):249–62. doi:10.1007/s10555-017-9673-1.; He A.D., Xie W., Song W., Ma Y.Y., Liu G., Liang M.L., Da X.W., Yao G.Q., Zhang B.X., Gao C.J., Xiang J.Z., Ming Z.Y. Platelet releasates promote the proliferation of hepatocellular carcinoma cells by suppressing the expression of KLF6. Sci Rep. 2017;7(1):3989. doi:10.1038/s41598-017-02801-1.; Banskota S., Gautam J., Regmi S.C., Gurung P., Park M.H., Kim S.J., Nam T.G., Jeong B.S., Kim J.A. BJ-1108, a 6-Amino-2,4,5- Trimethylpyridin-3-ol Analog, Inhibits Serotonin-Induced Angiogenesis and Tumor Growth through PI3K/NOX. Pathway. PLoS One. 2016;11(1):e0148133. doi:10.1371/journal.pone.0148133.; Wang S., Li Z., Xu R. Human Cancer and Platelet Interaction, a Potential Therapeutic Target. Int J Mol Sci. 2018;19(4):1246. doi:10.3390/ijms19041246.; https://journal.nodgo.org/jour/article/view/1040
-
4Academic Journal
المؤلفون: E. M. Golovina, K. S. Aslanyan, P. A. Zharkov, Е. М. Головина, К. С. Асланян, П. А. Жарков
المساهمون: The study was performed without external funding., Исследование проведено без спонсорской поддержки.
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 11, № 2 (2024); 90-96 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 11, № 2 (2024); 90-96 ; 2413-5496 ; 2311-1267
مصطلحات موضوعية: кровотечение, vitamin K, children, coagulopathy, bleeding, витамин К, дети, коагулопатия
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/1044/913; Newman P., Shearer M.J. Vitamin K metabolism. Subcell Biochem. 1998;30:455–88. doi:10.1007/978-1-4899-1789-8_19.; Schulte R., Jordan L.C., Morad A., Naftel R.P., Wellons J.C. 3rd, Sidonio R. Rise in late onset vitamin K defi ciency bleeding in young infants because of omission or refusal of prophylaxis at birth. Pediatr Neurol. 2014;50(6):564–8. doi:10.1016/j.pediatrneurol.2014.02.013.; Shearer M.J. Vitamin K metabolism and nutriture. Blood Rev. 1992;6:92–104. doi:10.1016/0268-960X(92)90011-E.; Ardell S., Off ringa M., Ovelman C., Soll R. Prophylactic vitamin K for the prevention of vitamin K defi ciency bleeding in preterm neonates. Cochrane Database Syst Rev. 2018;2(2):CD008342. doi:10.1002/14651858.CD008342.pub2.; Zipursky A. Prevention of vitamin K defi ciency bleeding in newborns. Br J Haematol. 1999;104:430–7. doi:10.1046/j.1365-2141.1999.01104.x.; Araki S., Shirahata A. Vitamin K Defi ciency Bleeding in Infancy. Nutrients. 2020;12(3):780. doi:10.3390/nu12030780.; Cornelissen M., von Kries R., Loughnan P., Schubiger G. Prevention of vitamin K defi ciency bleeding: Effi cacy of diff erent multiple oral dose schedules of vitamin K. Eur J Pediatr. 1997;156:126–30. doi:10.1007/s004310050570.; Jullien S. Vitamin K prophylaxis in newborns. BMC Pediatr. 2021;21(Suppl 1):350. doi:10.1186/s12887-021-02701-4.; Sankar M.J., Chandrasekaran A., Kumar P., Thukral A., Agarwal R., Paul V.K. Vitamin K prophylaxis for prevention of vitamin K defi ciency bleeding: a systematic review. J Perinatol. 2016;36 Suppl 1(Suppl 1):S29–35. doi:10.1038/jp.2016.30.; Higuera-Cetina C.I., Montaña-Gómez L.M., Picón-Jaimes Y.A., Orozco-Chinome J.E., Lozada-Martínez I.D., Moscote-Salazar L.R. Late-Onset Vitamin K Defi ciency Bleeding: A Preventable Pathology. J Pediatr Hematol Oncol. 2021;43(8):e1272–3. doi:10.1097/MPH.0000000000002315.; Townsend C.W. The haemorrhagic disease of the newborn. Arch Paediatr. 1894;11:559–565.; Dam H. Vitamin K, its discovery, biochemistry and application in medicine. J Mt Sinai Hosp N Y. 1946;12:961–70. PMID: 21011986.; World Health Organization. WHO recommendations on newborn health. Guidelines approved by the WHO guidelines review committee. Geneva, 2017. [Electronic resource]: https://apps.who.int/iris/bitstream/handle/10665/259269/WHO-MCA-17.07-eng.pdf;jsessionid=CF264F9448EAE361B5BE63F6B8BD284B?sequence=1 (appeal date 2024 Feb 1).; World Health Organization. Recommendations for management of common childhood conditions: evidence for technical update of pocket book recommendations, 2012. [Electronic resource]: https://apps.who.int/iris/bitstream/handle/10665/44774/9789241502825_eng.pdf?sequence=1 (appeal date 2024 Feb 1).; Диагностика и лечение геморрагической болезни новорожденных. Клинические рекомендации (проект). Ассоциация неонатологов, 2015. [Электронный ресурс]: https://babymed73.ru/upload/medialibrary/5ff/5ff 906e0c97d269f111098cb19299625.pdf.; Федеральные клинические рекомендации по диагностике и лечению геморрагической болезни новорожденных (проект). ФГБУ «ФНКЦ ДГОИ им. Дмитрия Рогачева» Минздрава России, Национальное общество детских гематологов и онкологов, 2014. [Электронный ресурс]: http://nodgo.org/sites/default/files/ФКР%20по%20диагностике%20и%20лечению%20геморрагической%20болезни%20новорожденных.pdf.; Неонатология. Национальное руководство в 2 т. Под ред. Н.Н. Володина, Д.Н. Дегтярева. М.: ГЭОТАР-Медиа, 2023. 752 с.; Chawla D., Deorari A.K., Saxena R., Paul V.K., Agarwal R., Biswas A., Meena A. Vitamin K1 versus vitamin K3 for prevention of subclinical vitamin defi ciency: a randomized controlled trial. Indian Pediatr. 2007;44(11):817–22. PMID: 18057477.; Dyggve H.V., Dam H., Søndergaard E. Comparison of the action of vitamin K1 with that of synkavit in the newborn. Acta Paediatr (Stockh). 1954;43(1):2–31. doi:10.1111/j.1651-2227.1954.tb03995.x.; Takahashi D., Shirahata A., Itoh S., Takahashi Y., Nishiguchi T., Matsuda Y. Vitamin K prophylaxis and late vitamin K defi ciency bleeding in infants: fi fth nationwide survey in Japan. Pediatr Int. 2011;53(6):897–901. doi:10.1111/j.1442-200X.2011.03392.x.; Ahmed S., Saikia R., Majumdar S., Banerjee Barua P., Priyadarshinee J., Paul S.P. Vitamin K defi ciency-related late-onset haemorrhagic disease of a newborn with acute subdural haemorrhage: long-term outcome. Sudan J Paediatr. 2021;21(2):195–9. doi:10.24911/SJP.106-1604579538.; Hasbaoui B.E., Karboubi L., Benjelloun B.S. Newborn haemorrhagic disorders: about 30 cases. Pan Afr Med J. 2017;28:150. doi:10.11604/pamj.2017.28.150.13159.; Демография. Федеральная служба государственной статистики. [Электронный ресурс]: https://rosstat.gov.ru/folder/12781 (дата обращения: 20.02.2024).; Mariani G., Bernardi F. Factor VII defi ciency. Semin Thromb Hemost. 2009;35(4):400–6. PMID: 20391303.; Menegatti M., Peyvandi F. Factor X Defi ciency. Semin Thromb Hemost. 2009;35(4):407–15. PMID: 19598069.; Lancellotti S., De Cristofaro R. Congenital prothrombin defi ciency. Semin Thromb Hemost. 2009;35(4):367–81. PMID: 19598065.; https://journal.nodgo.org/jour/article/view/1044
-
5Academic Journal
المؤلفون: T. Yu. Yafoshkina, Yu. A. Shifrin, D. V. Fedorova, D. B. Florinskiy, D. A. Evstratov, P. A. Zharkov, Т. Ю. Яфошкина, Ю. А. Шифрин, Д. В. Федорова, Д. Б. Флоринский, Д. А. Евстратов, П. А. Жарков
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 10, № 1 (2023); 33-40 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 10, № 1 (2023); 33-40 ; 2413-5496 ; 2311-1267
مصطلحات موضوعية: новые пероральные антикоагулянты, thrombosis therapy, thrombosis in children, anticoagulants, new oral anticoagulants, терапия тромбозов, тромбозы у детей, антикоагулянты
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/919/813; Raffini L., Huang Y.S., Witmer C., Feudtner C. Dramatic increase in venous thromboembolism in children’s hospitals in the United States from 2001 to 2007. Pediatrics. 2009;124(4):1001–8. doi:10.1542/peds.2009-0768.; Федеральные клинические рекомендации по диагностике, профилактике и лечению тромбозов у детей и подростков. М., 2015.; [Электронный ресурс]. URL: https://www.uptodate.com/contents/venous-thrombosis-and-thromboembolism-vte-in-children-riskfactors-clinical-manifestations-and-diagnosis. (Дата обращения: 14.09.2022).; De Crem N., Peerlinck K., Vanassche T., Vanheule K., Debaveye B., Middeldorp S., Verhamme P., Peetermans M. Abnormal uterine bleeding in VTE patients treated with rivaroxaban compared to vitamin K antagonists. Thromb Res. 2015;136(4):749–53. doi:10.1016/j.thromres.2015.07.030.; Kaatz S., Ahmad D., Spyropoulos A.C., Schulman S.; Subcommittee on Control of Anticoagulation. Definition of clinically relevant nonmajor bleeding in studies of anticoagulants in atrial fibrillation and venous thromboembolic disease in non-surgical patients: communication from the SSC of the ISTH. J Thromb Haemost. 2015;13(11):2119–26. doi:10.1111/jth.13140.; [Электронный ресурс]. URL: https://grls.rosminzdrav.ru/Grls_View_v2.aspx?routingGuid=073cdf44-10f6-4fa4-ab24-a5727d8c90f5. (Дата обращения: 17.01.2023).; Halton J., Brandão L.R., Luciani M., Bomgaars L., Chalmers E., Mitchell L.G., Nurmeev I., Sharathkumar A., Svirin P., Gorbatikov K., Tartakovsky I., Simetzberger M., Huang F., Sun Z., Kreuzer J., Gropper S., Reilly P., Brueckmann M., Albisetti M.; DIVERSITY Trial Investigators. Dabigatran etexilate for the treatment of acute venous thromboembolism in children (DIVERSITY): a randomised, controlled, open-label, phase 2b/3, non-inferiority trial. Lancet Haematol. 2021;8(1):e22–33. doi:10.1016/S2352-3026(20)30368-9.; Male C., Lensing A.W.A., Palumbo J.S., Kumar R., Nurmeev I., Hege K., Bonnet D., Connor P., Hooimeijer H.L., Torres M., Chan A.K.C., Kenet G., Holzhauer S., Santamaría A., Amedro P., Chalmers E., Simioni P., Bhat R.V., Yee D.L., Lvova O., BeyerWestendorf J., Biss T.T., Martinelli I., Saracco P., Peters M., Kállay K., Gauger C.A., Massicotte M.P., Young G., Pap A.F., Majumder M., Smith W.T., Heubach J.F., Berkowitz S.D., Thelen K., Kubitza D., Crowther M., Prins M.H., Monagle P.; EINSTEIN-Jr Phase 3 Investigators. Rivaroxaban compared with standard anticoagulants for the treatment of acute venous thromboembolism in children: a randomised, controlled, phase 3 trial. Lancet Haematol. 2020;7(1):e18–27. doi:10.1016/S2352-3026(19)30219-4.; Bryk A.H., Piróg M., Plens K., Undas A. Heavy menstrual bleeding in women treated with rivaroxaban and vitamin K antagonists and the risk of recurrent venous thromboembolism. Vascul Pharmacol. 2016;87:242–7. doi:10.1016/j.vph.2016.11.003.; Bannow B.S. Management of heavy menstrual bleeding on anticoagulation. Hematology Am Soc Hematol Educ Program. 2020;2020(1):533–7. doi:10.1182/hematology.2020000138.; Limperger V., Torge A., Kiesau B., Langer F., Kenet G., Mesters R., Juhl D., Stoll M., Shneyder M., Kowalski D., Bajorat T., Rocke A., Kuta P., Lasarow L., Spengler D., Junker R., Nowak-Göttl U. Validation of a predictive model for identifying an increased risk for recurrence in adolescents and young adults with a first provoked thromboembolism. Blood Cells Mol Dis. 2022;94:102651. doi:10.1016/j.bcmd.2022.102651.; Brandão L.R., Albisetti M., Halton J., Bomgaars L., Chalmers E., Mitchell L.G., Nurmeev I., Svirin P., Kuhn T., Zapletal O., Tartakovsky I., Simetzberger M., Huang F., Sun Z., Kreuzer J., Gropper S., Brueckmann M., Luciani M.; DIVERSITY Study Investigators. Safety of dabigatran etexilate for the secondary prevention of venous thromboembolism in children. Blood. 2020;135(7):491–504. doi:10.1182/blood.2019000998.; Pinchinat A., Otero N., Mahanti H., Morris E., Brudnicki A., Friedman D., Li S., Levendoglu-Tugal O., Cairo M.S.; A Pilot Study of an Oral Anticoagulant, Apixaban, in Secondary Prophylaxis of Venous Thromboembolism (VTE) in Children and Adolescents. Blood. 2019;134(Suppl. 1):2443. doi:10.1182/blood-2019-130468.; O’Brien S., Rodriguez V., Lew G., Newburger J., Schultz C., Orgel E., Derr K., Ranalli M., Esbenshade A., Hochberg J., Kang H., Dinikina Y., Crevar C., Donovan M., Dyme J., Favatella N., Mitchell L. PREVAPIX-ALL: Phase 3 Study of the Safety and Efficacy of Apixaban for Thromboprophylaxis versus Standard of Care in Newly Diagnosed Pediatric Acute Lymphoblastic Leukemia or Lymphoma (ALL/LL). [Электронный ресурс]. URL: https://abstracts.isth.org/abstract/prevapix-all-phase-3-study-of-the-safety-and-efficacy-ofapixaban-for-thromboprophylaxis-versus-standard-of-care-in-newlydiagnosed-pediatric-acute-lymphoblastic-leukemia-or-lymphoma-all-ll/. Accessed January 23, 2023.; McCrindle B.W., Michelson A.D., Van Bergen A.H., Horowitz E.S., Sandoval J.P., Justino H., Harris K.C., Jefferies J.L., Pina L.M., Peluso C., Nessel K., Lu W., Li J.S.; UNIVERSE Study Investigators. Thromboprophylaxis for children post-Fontan procedure: insights from the UNIVERSE study. J Am Heart Assoc. 2021;10(22):e021765. doi:10.1161/JAHA.120.021765.; Nowak-Göttl U., Junker R., Kreuz W., von Eckardstein A., Kosch A., Nohe N., Schobess R., Ehrenforth S.; Childhood Thrombophilia Study Group. Risk of recurrent venous thrombosis in children with combined prothrombotic risk factors. Blood. 2001;97(4):858–62. doi:10.1182/blood.v97.4.858.; Goldenberg N.A., Pounder E., Knapp-Clevenger R., Manco-Johnson M.J. Validation of upper extremity post-thrombotic syndrome outcome measurement in children. J. Pediatr. 2010;157(5):852–5. doi:10.1002/rth2.12251.; Goldenberg N.A., Donadini M.P., Kahn S.R., Crowther M., Kenet G., Nowak-Göttl U., Manco-Johnson M.J. Post-thrombotic syndrome in children: a systematic review of frequency of occurrence, validity of outcome measures, and prognostic factors. Haematologica. 2010;95(11):1952–9. doi:10.3324/haematol.2010.026989.; Avila M.L., Feldman B.M., Pullenayegum E., Lumia C., Montoya M.I., Vincelli J., Williams S., Brandão L.R. Post-thrombotic syndrome in children: Measurement properties of CAPTSure, a new diagnostic tool. Res Pract Thromb Haemost. 2019;3(4):652–7. doi:10.1002/rth2.12251.; https://journal.nodgo.org/jour/article/view/919
-
6Academic Journal
المؤلفون: T. Yu. Yafoshkina, P. A. Zharkov, Т. Ю. Яфошкина, П. А. Жарков
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 9, № 1 (2022); 45-51 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 9, № 1 (2022); 45-51 ; 2413-5496 ; 2311-1267
مصطلحات موضوعية: венозные тромбозы у детей, thrombosis, complications of deep vein thrombosis, venous thrombosis in children, тромбоз, осложнения тромбоза глубоких вен
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/801/718; Betensky M., Goldenberg N.A. Post-thrombotic syndrome in children. Thromb Res. 2018;164:129–35. doi:10.1016/j.thromres.2017.07.024.; Жарков П.А., Румянцев А.Г., Новичкова Г.А. Венозные тромбозы у детей со злокачественными новообразованиями (обзор литературы). Российский журнал детской гематологии и онкологии. 2015;2(1):66–74. doi:10.17650/2311-1267-2015-1-66-74.; Rajpurkar M., Sharathkumar A., Williams S., Lau K., Ling S.C., Chan A.K.C., Pediatric/Neonatal Hemostasis and Thrombosis Scientific and Standardization Subcommittee of the International Society on Thrombosis and Haemostasis. Recommendations for the assessment of non-extremity venous thromboembolism outcomes: communication from the SSC of the ISTH. J Thromb Haemost. 2015;13:477– 80. doi:10.1111/jth.12809.; Goldenberg N.A., Brandao L.R., Journeycake J., Kahn S.R., Monagle P., Revel-Vilk S., Sharathkumar A., Chan A.K.C. Defi nition of post-thrombotic syndrome following lower extremity deep venous thrombosis and standardization of outcome measurement in pediatric clinical investigations. J Thromb Haemost. 2012;10:477–80. doi:10.1111/j.1538-7836.2011.04594.x.; Kumar R., Rodriguez V., Matsumoto J.M., Khan S.P., Weaver A.L., McBane R.D., Beebe T.J., Heit J.A. Health-related quality of life in children and young adults with post-thrombotic syndrome: results from a cross-sectional study. Pediatr Blood Cancer. 2014;61(3):546–51. doi:10.1002/pbc.24840.; Johnson B.F., Manzo R.A., Bergelin R.O., Strandness D.E. Jr. Relationship between changes in deep venous system and the development of the post-thrombotic syndrome after an acute episode of lower limb vein thrombosis: a one-to-six year follow-up. J Vasc Surg. 1995;21(2):307–12; discussion 313. doi:10.1016/s0741-5214(95)70271-7.; Wakefield T.W., Henke P.K. The role of inflammation in early and late venous thrombosis: are there clinical implications? Semin Vasc Surg. 2005;18(3):118–29. doi:10.1053/j.semvascsurg.2005.05.003.; Shbaklo H., Holcroft C.A., Kahn S.R. Levels of inflammatory markers and the development of the post-thrombotic syndrome. Thromb Haemost. 2009;101(3):505. doi:10.1160/TH08-08-0511.; Roumen-Klappe E.M., den Heijer M., van Uum S.H., van der Ven-Jongekrijg J., van der Graaf F., Wollersheim H. Inflammatory response in the acute phase of deep venous thrombosis. J Vasc Surg. 2002;35(4):701–6. doi:10.1067/mva.2002.121746.; Meissner M.H., Zierler B.K., Bergelin R.O., Chandler W.L., Strandness D.E. Jr. Coagulation, fibrinolysis, and recanalization after acute deep venous thrombosis. J Vasc Surg. 2002;35(2):278–85. doi:10.1067/mva.2002.121066.; Varma M.R., Varga A.J., Knipp B.S., Sukheepod P., Upchurch G.R., Kunkel S.L., Wakefield T.W., Henke P.K. Neutropenia impairs venous thrombosis resolution in the rat. J Vasc Surg. 2003;38(5):1090–8. doi:10.1016/S0741-5214(03)00431-2.; Humphries J., McGuinness C.L., Smith A., Waltham M., Poston R., Burnand K.G. Monocyte chemotactic protein-1 (MCP-1) accelerates the organization and resolution of venous thrombi. J Vasc Surg. 1999;30(5):894–9. doi:10.1016/s0741-5214(99)70014-5.; Chandrashekar A., Garry J., Gasparis A., Labropoulos N. Vein wall remodeling in patients with acute deep vein thrombosis and chronic post-thrombotic changes. J Thromb Haemost. 2017;15(10):1989. doi:10.1111/jth.13793.; OʼShaughnessy A.M., Fitzgerald D.E. The patterns and distribution of residual abnormalities between the individual proximal venous segments after an acute deep vein thrombosis. J Vasc Surg. 2001;33(2):379–84. doi:10.1067/mva.2001.111983.; Singh H., Masuda E.M. Comparing short-term outcomes of femoral-popliteal and iliofemoral deep venous thrombosis: early lysis and development of reflux. Ann Vasc Surg. 2005;19(1):74. doi:10.1007/s10016-004-0133-3.; Meissner M.H., Manzo R.A., Bergelin R.O., Markel A., Strandness D.E. Jr. Deep venous insufficiency: the relationship between lysis and subsequent reflux. J Vasc Surg. 1993;18(4):596–605; discussion 606–8. PMID: 8411467.; Kreuz W., Stoll M., Junker R., Heinecke A., Schobess R., Kurnik K., Kelsch R., Nowak-Göttl U. Familial elevated factor VIII in children with symptomatic venous thrombosis and post-thrombotic syndrome: results of a multicenter study. Arterioscler Thromb Vasc Biol. 2006;26(8):1901–6. doi:10.1161/01.ATV.0000227510.36653.ed.; Kumar R., Rodriguez V., Matsumoto J.M., Khan S.P., Weaver A.L., McBane R.D., Beebe T.J., Heit J.A. Prevalence and risk factors for post-thrombotic syndrome after deep vein thrombosis in children: a cohort study. Thromb Res. 2015;135(2):347–51. doi:10.1016/j.thromres.2014.12.005.; Sharathkumar A.A., Pipe S.W. Post-thrombotic syndrome in children: a single center experience. J Pediatr Hematol Oncol. 2008;30(4):261–6. doi:10.1097/MPH.0b013e318162bcf5.; Luceri M.J., Tala J.A., Weismann C.G., Silva C.T., Faustino E.V.S. Prevalence of post-thrombotic syndrome after cardiac catheterization. Pediatr Blood Cancer. 2015;62(7):1222–7. doi:10.1002/pbc.25438.; Journeycake J.M., Buchanan G.R. Thrombotic complications of central venous catheters in children. Curr Opin Hematol. 2003;10(5):369–74. doi:10.1097/00062752-200309000-00008.; Revel-Vilk S., Menahem M., Stoffer C., Weintraub M. Post-thrombotic syndrome after central venous catheter removal in childhood cancer survivors is associated with a history of obstruction, Pediatr Blood Cancer. 2010;55(1):153–6. doi:10.1002/pbc.22459.; Polen E., Weintraub M., Stoffer C., Jaffe D.H., Burger A., Revel-Vilk S. Post-thrombotic syndrome after central venous catheter removal in childhood cancer survivors: a prospective cohort study. Pediatr Blood Cancer. 2015;62(2):285–90. doi:10.1002/pbc.25302.; Avila M.L., Pullenayegum E., Williams S., Yue N., Krol P., Brandão L.R. Post-thrombotic syndrome and other outcomes of lower extremity deep vein thrombosis in children. Blood. 2016;128(14):1862–9. doi:10.1182/blood-2016-03-704585.; Kuhle S., Koloshuk B., Marzinotto V., Bauman M., Massicotte P., Andrew M., Chan A., Abdolell M., Mitchell L. A cross-sectional study evaluating post-thrombotic syndrome in children. Thromb Res. 2003;111(4–5):227–33. doi:10.1016/j.thromres.2003.09.008.; Ginsberg J.S., Hirsh J., Julian J., Laande V., Vries M., Magier D., MacKinnon B., Gent M. Prevention and treatment of post-phlebitic syndrome: results of a 3-part study. Arch Intern Med. 2001;161(17):2105. doi:10.1001/archinte.161.17.2105.; Wille-Jоrgensen P., Jorgensen L.N., Crawford M. Asymptomatic postoperative deep vein thrombosis and the development of post-thrombotic syndrome. A systematic review and meta-analysis. Thromb Haemost. 2005;93(2):236. doi:10.1160/TH04-09-0570.; Kuhle S., Spavor M., Massicotte P., Halton J., Cherrick I., Dix D., Mahoney D., Bauman M., Desai S., Mitchell L.G. Prevalence of post-thrombotic syndrome following asymptomatic thrombosis in survivors of acute lymphoblastic leukemia. J Thromb Haemost. 2008;6(4):589–94. doi:10.1111/j.1538-7836.2008.02901.; Goldenberg N.A., Knapp-Clevenger R., Manco-Johnson M.J.; Mountain States Regional Thrombophilia Group. Elevated plasma factor VIII and D-dimer levels as predictors of poor outcomes of thrombosis in children. N Eng J Med. 2004;351(11):1081–8. doi:10.1056/NEJMoa040161.; Lyle C.A., Gibson E., Lovejoy A.E., Goldenberg N.A. Acute prognostic factors for post-thrombotic syndrome in children with limb DVT: a bi-institutional cohort study. Thromb Res. 2013;131(1):37–41. doi:10.1016/j.thromres.2012.10.012.; Ziegler S., Schillinger M., Maca T.H., Minar E. Post-thrombotic syndrome after primary event of deep venous thrombosis 10 to 20 years ago. Thromb Res. 2001;101(2):23–33. doi:10.1182/bloodadvances.2021004727.; van Dongen C.J., Prandoni P., Frulla M., Marchiori A., Prins M.H., Hutten B.A. Relation between quality of anticoagulant treatment and the development of the post-thrombotic syndrome. J Thromb Haemost. 2005;3(5):939. doi:10.1111/j.1538-7836.2005.01333.x.; Prandoni P., Frulla M., Sartor D., Concolato A., Girolami A. Vein abnormalities and the post-thrombotic syndrome. J Thromb Haemost. 2005;3(2):401–2. doi:10.1111/j.1538-7836.2004.01106.x.; Vedantham S. Valvular dysfunction and venous obstruction in the post-thrombotic syndrome. Thromb Res. 2009;123(Suppl 4):S62–5. doi:10.1016/S0049-3848(09)70146-X.; Comerota A.J., Grewal N., Martinez J.T., Chen J.T., Disalle R., Andrews L., Sepanski D., Assi Z. Post-thrombotic morbidity correlates with residual thrombus following catheter-directed thrombolysis for iliofemoral deep vein thrombosis. J Vasc Surg. 2012;55(3):768–73. doi:10.1016/j.jvs.2011.10.032.; Kahn S.R., Comerota A.J., Cushman M., Evans N.S., Ginsberg J.S., Goldenberg N.A., Gupta D.K., Prandoni P., Vedantham S., Walsh M.E., Weitz J.I., American Heart Association Council on Peripheral Vascular Disease, Council on Clinical Cardiology, and Council on Cardiovascular and Stroke Nursing. The post-thrombotic syndrome: evidence-based prevention, diagnosis, and treatment strategies: a scientific statement from the American Heart Association. Circulation. 2014;130(18):1636–61. doi:10.1161/CIR.0000000000000130.; Rabinovich A., Cohen J.M., Cushman M., Wells P.S., Rodger M.A., Kovacs M.J., Anderson D.R., Tagalakis V., Lazo-Langner A., Solymoss S., Miron M.J., Yeo E., Smith R., Schulman S., Kassis J., Kearon C., Chagnon I., Wong T., Demers C., Hanmiah R., Kaatz S., Selby R., Rathbun S., Desmarais S., Opatrny L., Ortel T.L., Ginsberg J.S., Kahn S.R. Inflammation markers and their trajectories after deep venous thrombosis in relation to risk of post-thrombotic syndrome. J Thromb Haemost. 2015;13(3):398–408. doi:10.1111/jth.12814.; Kahn S.R. The post-thrombotic syndrome. Hematology Am Soc Hematol Educ Program. 2016;2016(1):413–8. doi:10.1182/asheducation-2016.1.413.; Goldenberg N.A., Durham J.D., Knapp-Clevenger R., Manco-Johnson M.J. A thrombolytic regimen for high-risk deep venous thrombosis may substantially reduce the risk of post-thrombotic syndrome in children. Blood. 2007;110(1):45–53. doi:10.1182/blood-2006-12-061234.; Goldenberg N.A., Pounder E., Knapp-Clevenger R., Manco-Johnson M.J. Validation of upper extremity post-thrombotic syndrome outcome measurement in children. J Pediatr. 2010;157 (5):852–5. doi:10.1016/j.jpeds.2010.07.003.; Goldenberg N.A., Donadini M.P., Kahn S.R., Crowther M., Kenet G., Nowak-Göttl U., Manco-Johnson M.J. Post-thrombotic syndrome in children: a systematic review of frequency of occurrence, validity of outcome measures, and prognostic factors. Haematologica. 2010;95(11):1952–9. doi:10.3324/haematol.2010.026989.; Kahn S.R. Measurement properties of the Villalta scale to defi ne and classify the severity of the post-thrombotic syndrome. J Thromb Haemost. 2009;7(5):884–8. doi:10.1111/j.1538-7836.2009.03339.x.; Raffini L., Davenport J., Bevilacqua L., Iosifescu S. Comparison of 3 post-thrombotic syndrome assessment scales demonstrates significant variability in children and adolescents with deep vein thrombosis. J Pediatr Hematol Oncol. 2015;37(8):611–5. doi:10.1097/MPH.0000000000000399.; https://journal.nodgo.org/jour/article/view/801
-
7Academic Journal
المؤلفون: D. B. Florinsky, A. V. Pshonkin, A. V. Poletaev, D. V. Fedorova, E. A. Seregina, E. V. Ursulenko, O. P. Tolmacheva, P. A. Zharkov, Д. Б. Флоринский, А. В. Пшонкин, А. В. Полетаев, Д. В. Фёдорова, Е. А. Серёгина, Е. В. Урсуленко, О. П. Толмачева, П. А. Жарков
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 8, № 4 (2021); 90-94 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 8, № 4 (2021); 90-94 ; 2413-5496 ; 2311-1267
مصطلحات موضوعية: дети, factor XIII deficiency, prophylactic treatment, factor XIII concentrate, children, дефицит XIII фактора, профилактика, концентрат XIII фактора
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/776/702; Dorgalaleh A., Naderi M., Hosseini M.S., Alizadeh S., Hosseini S., Tabibian S., Eshghi P. Factor XIII deficiency in Iran: a comprehensive review of the literature. Semin Thromb Hemost. 2015;41(3):323–9. doi:10.1055/s-0034-1395350.; Ichinose A., Hendrickson L.E., Fujikawa K., Davie E.W. Amino acid sequence of the a subunit of human factor XIII. Biochemistry. 1986;25(22):6900–6. doi:10.1021/bi00370a025.; Bakker E.N., Pistea A., Spaan J.A., Rolf T., de Vries C.J., van Rooijen N., Candi E., VanBavel E. Flowdependent remodeling of small arteries in mice deficient for tissue-type transglutaminase possible compensation by macrophage-derived factor XIII. Circ Res. 2006;99(1):86–92. doi:10.1161/01.RES.0000229657.83816.a7.; Muszbek L., Yee V.C., Hevessy Z. Blood coagulation factor XIII: structure and function. Thromb Res. 1999;94(5):271–305. doi:10.1016/s0049-3848(99)00023-7.; Ichinose A., Davie E.W. Characterization of the gene for the a subunit of human factor XIII (plasma transglutaminase), a blood coagulation factor. Proc Natl Acad Sci U S A. 1988;85(16):5829–33. doi:10.1073/pnas.85.16.5829.; Muszbek L., Bagoly Z., Cairo A., Peyvandi F. Novel aspects of factor XIII deficiency. Curr Opin Hematol. 2011;18(5):366–72. doi:10.1097/MOH.0b013e3283497e3e.; Komaromi I., Bagoly Z., Muszbek L. Factor XIII: novel structural and functional aspects. J Thromb Haemost. 2011;9(1):9–20. doi:10.1111/j.1538-7836.2010.04070.x.; Kohler H., Ichinose A., Seitz R., Ariens R., Muszbek L. Diagnosis and classification of factor XIII deficiencies. J Thromb Haemost. 2011;9(7):1404–6. PMID:22946956.; Hsieh L., Nugent D. Factor XIII deficiency. Haemophilia. 2008;14(6):1190–200. doi:10.1111/j.1365-2516.2008.01857.x.; Mumford A.D., Ackroyd S., Alikhan R., Bowles L., Chowdary P., Grainger J., Mainwaring J., Mathias M., O’Connell N., BCSH Committee. Guideline for the diagnosis and management of the rare coagulation disorders: a United Kingdom Haemophilia Centre Doctors’ Organization guideline on behalf of the British Committee for Standards in Haematology, Br J Haematol. 2014;167(3):304–26. doi:10.1111/bjh.13058.; Dorgalaleh A., Rashidpanah J. Blood coagulation factor XIII and factor XIII deficiency. Blood Rev. 2016;30(6):461–75. PMID:27344554.; Caudill J.S., Nichols W.L., Plumhoff E.A., Schulte S.L., Winters J.L., Gastineau D.A., Rodriguez V. Comparison of coagulation factor XIII content and concentration in cryoprecipitate and fresh-frozen plasma. Transfusion. 2009;49(4):765–70. PMID:19192257.; Lusher J., Pipe S.W., Alexander S., Nugent D. Prophylactic therapy with Fibrogammin® P is associated with a decreased incidence of bleeding episodes: a retrospective study. Haemophilia. 2010;16:316–21. PMID:20017752.; Karaman S., Akkaya E., Genc S., Bilgili F., Kendirci A.S., Tugcu D., Unuvar A., Karakas Z., Demirkol D., Bayramoglu Z., Omer B. Congenital Factor XIII Deficiency With the Presence of Inhibitor: A Case Study. J Pediatr Hematol Oncol. 2021;43(1):e99–102. doi:10.1097/MPH.0000000000001671.; Mangla A., Hamad H., Kumar A. Factor XIII Deficiency. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2021 Jan. 2021 Oct 10. PMID:32491399.; Lassila R. Clinical Use of Factor XIII Concentrates. Semin Thromb Hemost. 2016;42(4):440–4. doi:10.1055/s-0036-1572324.; Carcao M., Altisent C., Castaman G., Fukutake K., Kerlin B.A., Kessler C., Lassila R., Nugent D., Oldenburg J., Garly M.L., Rosholm A., Inbal A. Recombinant FXIII (rFXIII-A2) Prophylaxis Prevents Bleeding and Allows for Surgery in Patients with Congenital FXIII A-Subunit Deficiency. Thromb Haemost. 2018;118(3):451–60. doi:10.1055/s-0038-1624581.; Государственный реестр лекарственных средств. [Электронный ресурс]: http://grls.rosminzdrav.ru/Default.aspx (дата обращения 20.10.2021).; Nascimento B., Goodnough L.T., Levy J.H. Cryoprecipitate therapy. Br J Anaesth. 2014;113(6):922–34. doi:10.1093/bja/aeu158.; Nugent D. Corifact™/Fibrogammin® P in the prophylactic treatment of hereditary factor XIII deficiency: results of a prospective, multicenter, open-label study. Thromb Res. 2012;130 Suppl 2:S12-4. doi:10.1016/S0049-3848(13)70005-7. PMID:23439001.; https://journal.nodgo.org/jour/article/view/776
-
8Academic Journal
المؤلفون: T. Yu. Yafoshkina, D. V. Fedorova, A. V. Poletaev, E. A. Seregina, P. A. Zharkov, Т. Ю. Яфошкина, Д. В. Федорова, А. В. Полетаев, Е. А. Серёгина, П. А. Жарков
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 9, № 3 (2022); 75-82 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 9, № 3 (2022); 75-82 ; 2413-5496 ; 2311-1267
مصطلحات موضوعية: тромбозы у детей, thrombophilia in children, heredity, thrombosis, venous thrombosis, thrombosis in children, тромбофилия у детей, наследственность, тромбоз, венозные тромбозы
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/860/758; Raffini L., Huang Y.S., Witmer C., Feudtner C. Dramatic increase in venous thromboembolism in children's hospitals in the United States from 2001 to 2007. Pediatrics. 2009;124(4):1001–8. doi:10.1542/peds.2009-0768.; [Электронный ресурс]. URL: https://www.uptodate.com/thrombophilia-testing-in-children-and-adolescents (Дата обращения: 14.09.2022).; Reitsma P.H., Rosendaal F.R. Past and future of genetic research in thrombosis. J Thromb Haemost. 2007;5 Suppl 1:264–9. doi:10.1111/j.1538-7836.2007.02502.x.; Жарков П.А. Влияние носительства протромботических полиморфизмов на риск развития тромбозов у детей. Автореф. дис. … канд. мед. наук. М., 2013.; Revel-Vilk S., Chan A., Bauman M., Massicotte P. Prothrombotic conditions in an unselected cohort of children with venous thromboembolic disease. J Thromb Haemost. 2003;1(5):915–21. doi:10.1046/j.1538-7836.2003.00158.x.; Клинические рекомендации. Детская гематология. Под ред. Жуковской Е.В., Румянцева А.Г., Масчана А.А. М.: ГЭОТАР-Медиа, 2015. 656 с.; [Электронный ресурс]. URL: https://www.uptodate.com/contents/antithrombin-deficiency (Дата обращения: 14.09.2022).; [Электронный ресурс]. URL: https://www.uptodate.com/contents/protein-s-deficiency (Дата обращения: 14.09.2022).; [Электронный ресурс]. URL: https://www.uptodate.com/contents/protein-c-deficiency (Дата обращения: 14.09.2022).; Kamphuisen P.W., Eikenboom J.C., Bertina R.M. Elevated factor VIII levels and the risk of thrombosis. Arterioscler Thromb Vasc Biol. 2001;21(5):731–8. doi:10.1161/01.ATV.21.5.731.; [Электронный ресурс]. URL: https://www.uptodate.com/contents/overview-ofhomocysteine/abstract/81 (Дата обращения 14.09.2022).; Nowak-Göttl U., Junker R., Hartmeier M., Koch H.G., Munchow N., Assmann G., von Eckardstein A. Increased lipoprotein (a) is an important risk factor for venous thromboembolism in childhood. Circulation. 1999;100(7):743–8. doi:10.1161/01.cir.100.7.743.; Miyakis S., Lockshin M.D., Atsumi T., Branch D.W., Brey R.L., Cervera R., Groot P.G., Koike T., Meroni P.L., Reber G., Shoenfeld Y., Tincani A., Vlachoyiannopolos P.G., Krilis S.A. International consensus statement on an update of the classification criteria for defi nite antiphospholipid syndrome (APS). J Thromb Haemost. 2006;4(2):295–306. doi:10.1111/j.1538-7836.2006.01753.x.; Tuckuviene R., Christensen A.L., Helgestad J., Johnsen S.P., Kristensen S.R. Pediatric venous and arterial noncerebral thromboembolism in Denmark: a nationwide population-based study. J Pediatr. 2011;159(4):663–9. doi:10.1016/j.jpeds.2011.03.052.; Takemoto C.M., Sohi S., Desai K., Bharaj R., Khanna A., McFarland S., Klaus S., Irshad A., Goldenberg N.A., Strouse J.J., Streiff M.B. Hospital-associated venous thromboembolism in children: incidence and clinical characteristics. J Pediatr. 2014;164(2):332–8. doi:10.1016/j.jpeds.2013.10.025.; Van Ommen C.H., Heijboer H., van den Dool E.J., Hutten B.A., Peters M. Pediatric venous thromboembolic disease in one single center: congenital prothrombotic disorders and the clinical outcome. J Thromb Haemost. 2003;1(12):2516–22. doi:10.1046/j.1538-7836.2003.00465.x.; Mitchell L.G., Andrew M., Hanna K., Abshire T., Halton J., Anderson R., Cherrick I., Desai S, Mahoney D., McCuster P., Wu J., Dahl G., Chait P., de Veber G., Lee K.-J., Mikulis D., Ginsberg J., Way C., Prophylactic Antithrombin Replacement in Kids with Acute Lymphoblastic Leukemia Treated with Asparaginase Group (PARKAA). A prospective cohort study determining the prevalence of thrombotic events in children with acute lymphoblastic leukemia and a central venous line who are treated with L-asparaginase: results of the Prophylactic Antithrombin Replacement in Kids with Acute Lymphoblastic Leukemia Treated with Asparaginase (PARKAA) Study. Cancer. 2003;97(2):508–16. doi:10.1002/cncr.11042.; Raffini L. Thrombophilia in Children: Who to Test, How, When, and Why? Hematology Am Soc Hematol Educ Program. 2008;228–35. doi:10.1182/asheducation-2008.1.228.; Turebylu R., Salis R., Erbe R., Martin D., Lakshminrusimha S., Ryan R.M. Genetic prothrombotic mutations are common in neonates but are not associated with umbilical catheter-associated thrombosis. J Perinatol. 2007;27(8):490–5. doi:10.1038/sj.jp.7211786.; Nowak-Gottl U., Dubbers A., Kececioglu D., Koch H.G., Kotthoff S., Runde J., Vielhaber H. Factor V Leiden, protein C, and lipoprotein (a) in catheter-related thrombosis in childhood: a prospective study. J Pediatr. 1997;131(4):608–12. doi:10.1016/s0022-3476(97)70071-4.; Kenet G., Limperger V., Shneyder M., Nowak-Göttl U. Risk factors for symptomatic venous and arterial thromboembolism in newborns, children and adolescents – What did we learn within the last 20 years? Blood Cells Mol Dis. 2017;67:18–22. doi:10.1016/j.bcmd.2016.12.003.; Neshat-Vahid S., Pierce R., Hersey D., Raffi ni L.J., Faustino E.V. Association of thrombophilia and catheter-associated thrombosis in children: a systematic review and meta-analysis. J Thromb Haemost. 2016;14(9):1749. doi:10.1111/jth.13388.; Han H., Hensch L., Hui S.R., Teruya J. Evaluation and Management of Coagulopathies and Thrombophilias in Pediatric Patients. Clin Lab Med. 2021;41(1):83–100. doi:10.1016/j.cll.2020.10.006.; [Электронный ресурс]. URL: https://www.uptodate.com/venous-thrombosis-and-thromboembolism-vte-in-children-risk-factors-clinical-manifestations-and-diagnosis (Дата обращения: 14.09.2022).; [Электронный ресурс]. URL: https://www.uptodate.com/evaluating-adult-patients-with-established-venous-thromboembolism-for-acquired-and-inherited-risk-factors (Дата обращения: 14.09.2022).; https://journal.nodgo.org/jour/article/view/860
-
9Academic Journal
المؤلفون: P. A. Zharkov, П. А. Жарков
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 9, № 3 (2022); 56-64 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 9, № 3 (2022); 56-64 ; 2413-5496 ; 2311-1267
مصطلحات موضوعية: дети, inhibitors, immune tolerance induction, treatment, factor, children, ингибитор, индукция иммунной толерантности, лечение, фактор
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/858/756; Srivastava A., Santagostino E., Dougall A., Kitchen S., Sutherland M., Pipe S.W., Carcao M., Mahlangu J., Ragni M.V., Windyga J., Llinás A., Goddard N.J., Mohan R., Poonnoose P.M., Feldman B.M., Lewis S.Z., van den Berg H.M., Pierce G.F. WFH Guidelines for the Management of Hemophilia, 3 rd edition. Haemophilia. 2020;26:1–158. doi:10.1111/hae.14046.; Aledort L., Mannucci P.M., Schramm W., Tarantino M. Factor VIII replacement is still the standard of care in haemophilia A. Blood Transfus. 2019;17(6):479–86. doi:10.2450/2019.0211-19.; Peyvandi F., Mannucci P.M., Garagiola I., El-Beshlawy A., Elalfy M., Ramanan V., Eshghi P., Hanagavadi S., Varadarajan R., Karimi M., Manglani M.V., Ross C., Young G., Seth T., Apte S., Nayak D.M., Santagostino E., Mancuso M.E., Sandoval Gonzalez A.C., Mahlangu J.N., Bonanad Boix S., Cerqueira M., Ewing N.P., Male C., Owaidah T., Soto Arellano V., Kobrinsky N.L., Majumdar S., Perez Garrido R., Sachdeva A., Simpson M., Thomas M., Zanon E., Antmen B., Kavakli K., Manco-Johnson M.J., Martinez M., Marzouka E., Mazzucconi M.G., Neme D., Palomo Bravo A., Paredes Aguilera R., Prezotti A., Schmitt K., Wicklund B.M., Zulfi kar B., Rosendaal F.R. A Randomized Trial of Factor VIII and Neutralizing Antibodies in Hemophilia A. N Engl J Med. 2016;374(21):2054–64. doi:10.1056/NEJMoa1516437. PMID: 27223147.; Rota M., Cortesi P.A., Steinitz-Trost K.N., Reininger A.J., Gringeri A., Mantovani L.G. Meta-analysis on incidence of inhibitors in patients with haemophilia A treated with recombinant factor VIII products. Blood Coagul Fibrinolysis. 2017;28(8):627–37. doi:10.1097/MBC.0000000000000647. PMID: 28678027.; Wight J., Paisley S. The epidemiology of inhibitors in haemophilia A: a systematic review. Haemophilia. 2003;9(4):418–35. doi:10.1046/j.1365-2516.2003.00780.x. PMID: 12828678.; Hassan S., Cannavò A., Gouw S.C., Rosendaal F.R., van der Bom J.G. Factor VIII products and inhibitor development in previously treated patients with severe or moderately severe hemophilia A: a systematic review. J Thromb Haemost. 2018;16(6):1055–68. doi:10.1111/jth.14124.; Chai-Adisaksopha C., Nevitt S.J., Simpson M.L., Janbain M., Konkle B.A. Bypassing agent prophylaxis in people with hemophilia A or B with inhibitors. Cochrane Database Syst Rev. 2017;9(9):CD011441. doi:10.1002/14651858.CD011441.pub2.; Жарков П.А., Воронин К.А., Андреева Т.А., Асекретова Т.В., Белкина Ю.Е., Демихов В.Г., Зинина Е.Е., Колясина Т.А., Лебедев В.В., Маркова И.В., Осмульская Н.С., Петров В.Ю., Скобин В.Б., Спичак О.В., Шелехова Т.В., Шерстнев Д.Г. Значительное снижение количества кровотечений у детей с ингибиторной формой гемофилии А в реальной клинической практике применения эмицизумаба. Вопросы гематологии/онкологии и иммунопатологии в педиатрии. 2022;21(1):66–71. doi:10.24287/1726-1708-2022-21-1-66-71; O’Hara J., Sima C.S., Frimpter J., Paliargues F., Chu P., Presch I. Long-term outcomes from prophylactic or episodic treatment of haemophilia A: A systematic review. Haemophilia. 2018;24(5):e301–11. doi:10.1111/hae.13546.; Delgado-Flores C.J., García-Gomero D., Salvador-Salvador S., Montes-Alvis J., Herrera-Cunti C., Taype-Rondan A. Effects of replacement therapies with clotting factors in patients with hemophilia: A systematic review and meta-analysis. PLoS One. 2022;17(1):e0262273. doi:10.1371/journal.pone.0262273. PMID: 35030189.; Von Mackensen S., Kalnins W., Krucker J., Weiss J., Miesbach W., Albisetti M., Pabinger I., Oldenburg J. Haemophilia patients' unmet needs and their expectations of the new extended half-life factor concentrates. Haemophilia. 2017;23(4):566–74. doi:10.1111/hae.13221.; Hermans C., Mancuso M.E., Nolan B., Pasi K.J. Recombinant factor VIII Fc for the treatment of haemophilia A. Eur J Haematol. 2021;106(6):745–61. doi:10.1111/ejh.13610.; Siekmann J., Turecek P.L. PEGylation of human coagulation factor VIII and other plasma proteins. Polymer-Protein Conjugates. 2020:155–74. doi:10.1016/B978-0-444-64081-9.00008-5.; Raso S., Hermans C. Lonoctocog alfa (rVIII-SingleChain) for the treatment of haemophilia A. Expert Opin Biol Ther. 2018;18(1):87–94. doi:10.1080/14712598.2018.1416088.; Morfi ni M. Simoctocog alfa for the treatment of hemophilia A. Expert Opin Biol Ther. 2017;17(12):1573–80. doi:10.1080/14712598.2017.1391785.; Lissitchkov T., Klukowska A., Pasi J., Kessler C.M., Klamroth R., Liesner R.J., Belyanskaya L., Walter O., Knaub S., Bichler J., Jansen M., Oldenburg J. Efficacy and safety of simoctocog alfa (Nuwiq® ) in patients with severe hemophilia A: a review of clinical trial data from the GENA program. Ther Adv Hematol. 2019;10:2040620719858471. doi:10.1177/2040620719858471.; Di Minno M.N.D., Di Minno A., Calcaterra I., Cimino E., Dell’Aquila F., Franchini M. Enhanced Half-Life Recombinant Factor VIII Concentrates for Hemophilia A: Insights from Pivotal and Extension Studies. Semin Thromb Hemost. 2021;47(1):32–42. doi:10.1055/s-0040-1718887.; Klamroth R., Wojciechowski P., Aballéa S., Diamand F., Hakimi Z., Nazir J., Abad-Franch L., Lethagen S., Santagostino E., Tarantino M.D. Efficacy of rFVIIIFc versus Emicizumab for the Treatment of Patients with Hemophilia A without Inhibitors: Matching-Adjusted Indirect Comparison of A-LONG and HAVEN Trials. J Blood Med. 2021;12:115–22. doi:10.2147/JBM.S288283.; Konkle B.A., Shapiro A.D., Quon D.V., Staber J.M., Kulkarni R., Ragni M.V., Chhabra E.S., Poloskey S., Rice K., Katragadda S., Fruebis J., Benson C.C. BIVV001 Fusion Protein as Factor VIII Replacement Therapy for Hemophilia A. N Engl J Med. 2020;383(11):1018–27. doi:10.1056/NEJMoa2002699. PMID: 32905674.; Lenting P.J., Denis C.V., Christophe O.D. Emicizumab, a bispecific antibody recognizing coagulation factors IX and X: how does it actually compare to factor VIII? Blood. 2017;130(23):2463–8. doi:10.1182/blood-2017-08-801662.; Lauritzen B., Bjelke M., Björkdahl O., Bloem E., Keane K., Kjalke M., Rossen M., Lippert S.L., Weldingh K.N., Skydsgaard M., Kjellev S. A novel next-generation FVIIIa mimetic, Mim8, has a favorable safety profile and displays potent pharmacodynamic effects: Results from safety studies in cynomolgus monkeys. J Thromb Haemost. 2022;20(6):1312–24. doi:10.1111/jth.15682.; Lindley C.M., Sawyer W.T., Macik B.G., Lusher J., Harrison J.F., Baird-Cox K., Birch K., Glazer S., Roberts H.R. Pharmacokinetics and pharmacodynamics of recombinant factor VIIa. Clin Pharmacol Ther. 1994;55(6):638–48. doi:10.1038/clpt.1994.80. PMID: 8004880.; Wang M., Lawrence J.B., Quon D.V., Ducore J., Simpson M.L., Boggio L.N., Mitchell I.S., Yuan G., Alexander W.A., Schved J.F. PERSEPT 1: a phase 3 trial of activated eptacog beta for on-demand treatment of haemophilia inhibitor-related bleeding. Haemophilia. 2017;23(6):832–43. doi:10.1111/hae.13301.; Gruppo R.A., Malan D., Kapocsi J., Nemes L., Hay C.R.M., Boggio L., Chowdary P., Tagariello G., von Drygalski A., Hua F., Scaramozza M., Arkin S.; Marzeptacog alfa (activated) Study Group Investigators. Phase 1, single-dose escalating study of marzeptacog alfa (activated), a recombinant factor VIIa variant, in patients with severe hemophilia. J Thromb Haemost. 2018;16(10):1984–93. doi:10.1111/jth.14247.; Abildgaard C.F., Penner J.A., Watson-Williams E.J. Anti-inhibitor Coagulant Complex (Autoplex) for treatment of factor VIII inhibitors in hemophilia. Blood. 1980;56(6):978–84. PMID: 6777002.; Eichler H., Angchaisuksiri P., Kavakli K., Knoebl P., Windyga J., Jiménez-Yuste V., Harder Delff P., Chowdary P. Concizumab restores thrombin generation potential in patients with haemophilia: Pharmacokinetic/pharmacodynamic modelling results of concizumab phase 1/1b data. Haemophilia. 2019;25(1):60–6. doi:10.1111/hae.13627.; Pasi K.J., Lissitchkov T., Mamonov V., Mant T., Timofeeva M., Bagot C., Chowdary P., Georgiev P., Gercheva-Kyuchukova L., Madigan K., Van Nguyen H., Yu Q., Mei B., Benson C.C., Ragni M.V. Targeting of antithrombin in hemophilia A or B with investigational siRNA therapeutic fitusiran-Results of the phase 1 inhibitor cohort. J Thromb Haemost. 2021;19(6):1436–46. doi:10.1111/jth.15270.; Hamedani N.S., Rühl H., Zimmermann J.J., Heiseler T., Oldenburg J., Mayer G., Pötzsch B., Müller J. In Vitro Evaluation of Aptamer-Based Reversible Inhibition of Anticoagulant Activated Protein C as a Novel Supportive Hemostatic Approach. Nucleic Acid Ther. 2016;26(6):355–62. doi:10.1089/nat.2016.0645.; Polderdijk S.G., Adams T.E., Ivanciu L., Camire R.M., Baglin T.P., Huntington J.A. Design and characterization of an APC-specific serpin for the treatment of hemophilia. Blood. 2017;129(1):105–13. doi:10.1182/blood-2016-05-718635.; Zhao X.Y., Wilmen A., Wang D., Wang X., Bauzon M., Kim J.Y., Linden L., Li L., Egner U., Marquardt T., Moosmayer D., Tebbe J., Glück J.M., Ellinger P., McLean K., Yuan S., Yegneswaran S., Jiang X., Evans V., Gu J.M., Schneider D., Zhu Y., Xu Y., Mallari C., Hesslein A., Wang Y., Schmidt N., Gutberlet K., Ruehl-Fehlert C., Freyberger A., Hermiston T., Patel C., Sim D., Mosnier L.O., Laux V. Targeted inhibition of activated protein C by a non-active-site inhibitory antibody to treat hemophilia. Nat Commun. 2020;11(1):2992. doi:10.1038/s41467-020-16720-9.; Prince R., Bologna L., Manetti M., Melchiorre D., Rosa I., Dewarrat N., Suardi S., Amini P., Fernández J.A., Burnier L., Quarroz C., Reina Caro M.D., Matsumura Y., Kremer Hovinga J.A., Griffin J.H., Simon H.U., Ibba-Manneschi L., Saller F., Calzavarini S., Angelillo-Scherrer A. Targeting anticoagulant protein S to improve hemostasis in hemophilia. Blood. 2018;131(12):1360–71. doi:10.1182/blood-2017-09-800326.; Инструкция по медицинскому применению препарата Эмицизумаб. [Электронный ресурс] URL: https://www.rlsnet.ru/tn_index_id_96727 (дата обращения 10.2021).; Mahlangu J., Oldenburg J., Paz-Priel I., Negrier C., Niggli M., Mancuso M.E., Schmitt C., Jiménez-Yuste V., Kempton C., Dhalluin C., Callaghan M.U., Bujan W., Shima M., Adamkewicz J.I., Asikanius E., Levy G.G., Kruse-Jarres R. Emicizumab Prophylaxis in Patients Who Have Hemophilia A without Inhibitors. N Engl J Med. 2018;379(9):811–22. doi:10.1056/NEJMoa1803550. PMID: 30157389.; Young G., Liesner R., Chang T., Sidonio R., Oldenburg J., Jiménez-Yuste V., Mahlangu J., Kruse-Jarres R., Wang M., Uguen M., Doral M.Y., Wright L.Y., Schmitt C., Levy G.G., Shima M., Mancuso M.E. A multicenter, open-label phase 3 study of emicizumab prophylaxis in children with hemophilia A with inhibitors. Blood. 2019;134(24):2127–38. doi:10.1182/blood.2019001869.; Pipe S.W., Shima M., Lehle M., Shapiro A., Chebon S., Fukutake K., Key N.S., Portron A., Schmitt C., Podolak-Dawidziak M., Selak Bienz N., Hermans C., Campinha-Bacote A., Kiialainen A., Peerlinck K., Levy G.G., Jiménez-Yuste V. Efficacy, safety, and pharmacokinetics of emicizumab prophylaxis given every 4 weeks in people with haemophilia A (HAVEN 4): a multicentre, open-label, non-randomised phase 3 study. Lancet Haematol. 2019;6(6):e295–305. doi:10.1016/S2352-3026(19)30054-7.; Клинические рекомендации «Гемофилия» (утверждены Минздравом России), 2018 г. [Электронный ресурс] URL: https://legalacts.ru/doc/klinicheskie-rekomendatsii-gemofilija-utv-minzdravom-rossii/ [Clinical guidelines “Hemophilia” (approved by the Ministry of Health of Russia), 2018.; Watanabe A.H., Lee S.W.H., Chai-Adisaksopha C., Lim M.Y., Chaiyakunapruk N. Budget Impact of Emicizumab for Routine Prophylaxis of Bleeding Episodes in Patients With Hemophilia A With Inhibitors. Value Health Reg Issues. 2022;28:7–13. doi:10.1016/j.vhri.2021.07.003.; Samelson-Jones B.J., Guelcher C., Kuhn J., Butler R., Massey G., Guerrera M.F., Raffini L. Real-world cost estimates of initiating emicizumab in US patients with haemophilia A. Haemophilia. 2021;27(4):591–8. doi:10.1111/hae.14347.; Saiyarsarai P., Robabpour Derakhshan A., Khedmati J., Eshghi P., Seyedifar M. A comparison between on-demand usage of rFVIIa vs prophylaxis use of emicizumab in high titer inhibitory hemophilia A patients in Iran: A cost-utility analysis. Medicine (Baltimore). 2021;100(40):e27303. doi:10.1097/MD.0000000000027303.; Polack B., Trossaërt M., Cousin M., Baffert S., Pruvot A., Godard C. Cost-effectiveness of emicizumab vs bypassing agents in the prevention of bleeding episodes in haemophilia A patients with anti-FVIII inhibitors in France. Haemophilia. 2021;27(1):e1–11. doi:10.1111/hae.14129.; Cortesi P.A., Castaman G., Trifi rò G., Creazzola S.S., Improta G., Mazzaglia G., Molinari A.C., Mantovani L.G. Cost-Effectiveness and Budget Impact of Emicizumab Prophylaxis in Haemophilia A Patients with Inhibitors. Thromb Haemost. 2020;120(2):216–28. doi:10.1055/s-0039-3401822.; Lee H., Cho H., Han J.W., Kim A.Y., Park S., Lee M., Cho S., Baik D., Kang H.Y. Cost-utility analysis of emicizumab prophylaxis in haemophilia A patients with factor VIII inhibitors in Korea. Haemophilia. 2021;27(1):e12–21. doi:10.1111/hae.14143.; Schmitt C., Adamkewicz J.I., Xu J., Petry C., Catalani O., Young G., Negrier C., Callaghan M.U., Levy G.G. Pharmacokinetics and Pharmacodynamics of Emicizumab in Persons with Hemophilia A with Factor VIII Inhibitors: HAVEN 1 Study. Thromb Haemost. 2021;121(3):351–60. doi:10.1055/s-0040-1717114.; Yoneyama K., Schmitt C., Kotani N., Levy G.G., Kasai R., Iida S., Shima M., Kawanishi T. A Pharmacometric Approach to Substitute for a Conventional Dose-Finding Study in Rare Diseases: Example of Phase III Dose Selection for Emicizumab in Hemophilia A. Clin Pharmacokinet. 2018;57(9):1123–34. doi:10.1007/s40262-017-0616-3.; Ferrière S., Peyron I., Christophe O.D., Kawecki C., Casari C., Muczynski V., Nathwani A., Kauskot A., Lenting P.J., Denis C.V. A hemophilia A mouse model for the in vivo assessment of emicizumab function. Blood. 2020;136(6):740–8. doi:10.1182/blood.2019004334. PMID: 32369559.; Oldenburg J., Mahlangu J.N., Kim B., Schmitt C., Callaghan M.U., Young G., Santagostino E., Kruse-Jarres R., Negrier C., Kessler C., Valente N., Asikanius E., Levy G.G., Windyga J., Shima M. Emicizumab Prophylaxis in Hemophilia A with Inhibitors. N Engl J Med. 2017;377(9):809–18. doi:10.1056/NEJMoa1703068.; Hartmann R., Feenstra T., Valentino L., Dockal M., Scheiflinger F. In vitro studies show synergistic effects of a procoagulant bispecific antibody and bypassing agents. J Thromb Haemost. 2018. doi:10.1111/jth.14203.; Schultz N.H., Glosli H., Bjørnsen S., Holme P.A. The effect of emicizumab and bypassing agents in patients with hemophilia –An in vitro study. Res Pract Thromb Haemost. 2021;5(5):e12561. doi:10.1002/rth2.12561.; Kjalke M., Kjelgaard-Hansen M., Andersen S., Hilden I. Thrombin generation potential in the presence of concizumab and rFVIIa, APCC, rFVIII, or rFIX: In vitro and ex vivo analyses. J Thromb Haemost. 2021;19(7):1687–96. doi:10.1111/jth.15323.; Rota M., Cortesi P.A., Crea R., Gringeri A., Mantovani L.G. Thromboembolic event rate in patients exposed to anti-inhibitor coagulant complex: a meta-analysis of 40-year published data. Blood Adv. 2017;1(26):2637–42. doi:10.1182/bloodadvances.2017011536.; Rajpurkar M., Croteau S.E., Boggio L., Cooper D.L. Thrombotic events with recombinant activated factor VII (rFVIIa) in approved indications are rare and associated with older age, cardiovascular disease, and concomitant use of activated prothrombin complex concentrates (aPCC). J Blood Med. 2019;10:335–40. doi:10.2147/JBM.S219573.; Jiménez-Yuste V., Auerswald G., Benson G., Dolan G., Hermans C., Lambert T., Ljung R., Morfi ni M., Santagostino E., Zupančić Šalek S. Practical considerations for nonfactor-replacement therapies in the treatment of haemophilia with inhibitors. Haemophilia. 2021;27(3):340–50. doi:10.1111/hae.14167.; Batty P., Lillicrap D. Gene therapy for hemophilia: Current status and laboratory consequences. Int J Lab Hematol. 2021;43 Suppl 1:117–23. doi:10.1111/ijlh.13605.; Pasi K.J., Laffan M., Rangarajan S., Robinson T.M., Mitchell N., Lester W., Symington E., Madan B., Yang X., Kim B., Pierce G.F., Wong W.Y. Persistence of haemostatic response following gene therapy with valoctocogene roxaparvovec in severe haemophilia A. Haemophilia. 2021;27(6):947–56. doi:10.1111/hae.14391.; Hay C.R., Palmer B., Chalmers E., Liesner R., Maclean R., Rangarajan S., Williams M., Collins P.W.; United Kingdom Haemophilia Centre Doctors’ Organisation (UKHCDO). Incidence of factor VIII inhibitors throughout life in severe hemophilia A in the United Kingdom. Blood. 2011;117(23):6367–70. doi:10.1182/blood-2010-09-308668.; Delavenne X., Dargaud Y. Pharmacokinetics for haemophilia treaters: Meaning of PK parameters, interpretation pitfalls, and use in the clinic. Thromb Res. 2020;192:52–60. doi:10.1016/j.thromres.2020.05.005.; Young G., Sørensen B., Dargaud Y., Negrier C., Brummel-Ziedins K., Key N.S. Thrombin generation and whole blood viscoelastic assays in the management of hemophilia: current state of art and future perspectives. Blood. 2013;121(11):1944–50. doi:10.1182/blood-2012-08-378935.; Fischer K., Lassila R., Peyvandi F., Calizzani G., Gatt A., Lambert T., Windyga J., Iorio A., Gilman E., Makris M.; EUHASS participants. Inhibitor development in haemophilia according to concentrate. Four-year results from the European HAemophilia Safety Surveillance (EUHASS) project. Thromb Haemost. 2015;113(5):968–75. doi:10.1160/TH14-10-0826.; Gouw S.C., van den Berg H.M., Fischer K., Auerswald G., Carcao M., Chalmers E., Chambost H., Kurnik K., Liesner R., Petrini P., Platokouki H., Altisent C., Oldenburg J., Nolan B., Garrido R.P., Mancuso M.E., Rafowicz A., Williams M., Clausen N., Middelburg R.A., Ljung R., van der Bom J.G.; PedNet and Research of Determinants of INhibitor development (RODIN) Study Group. Intensity of factor VIII treatment and inhibitor development in children with severe hemophilia A: the RODIN study. Blood. 2013;121(20):4046–55. doi:10.1182/blood-2012-09-457036.; Oldenburg J., Jiménez-Yuste V., Peiró-Jordán R., Aledort L.M., Santagostino E. Primary and rescue immune tolerance induction in children and adults: a multicentre international study with a VWF-containing plasma-derived FVIII concentrate. Haemophilia. 2014;20(1):83–91. doi:10.1111/hae.12263.; Kreuz W., Escuriola Ettingshausen C., Vdovin V., Zozulya N., Plyushch O., Svirin P., Andreeva T., Bubanská E., Campos M., Benedik-Dolničar M., Jiménez-Yuste V., Kitanovski L., Klukowska A., Momot A., Osmulskaya N., Prieto M., Šalek S.Z., Velasco F., Pavlova A., Oldenburg J., Knaub S., Jansen M., Belyanskaya L., Walter O.; ObsITI study group; ObsITI committee. First prospective report on immune tolerance in poor risk haemophilia A inhibitor patients with a single factor VIII/von Willebrand factor concentrate in an observational immune tolerance induction study. Haemophilia. 2016;22(1):87–95. doi:10.1111/hae.12774.; Escuriola Ettingshausen C., Kreuz W. A review of immune tolerance induction with Haemate P in haemophilia A. Haemophilia. 2014;20(3):333–9. doi:10.1111/hae.12288.; Rothschild C., D’Oiron R., Borel-Derlon A., Gruel Y., Navarro R., Negrier C. Use of Haemate( ® ) P as immune tolerance induction in patients with severe haemophilia A who failed previous induction attempts: a multicentre observational study. Haemophilia. 2013;19(2):281–6. doi:10.1111/hae.12018.; Suzuki T., Arai M., Amano K., Kagawa K., Fukutake K. Factor VIII inhibitor antibodies with C2 domain specificity are less inhibitory to factor VIII complexed with von Willebrand factor. Thromb Haemost. 1996;76(5):749–54. PMID: 8950785.; Kallas A., Talpsep T. Von Willebrand factor in factor VIII concentrates protects against neutralization by factor VIII antibodies of haemophilia A patients. Haemophilia. 2001;7(4):375–80. doi:10.1046/j.1365-2516.2001.00530.x. PMID: 11442642.; Astermark J., Voorberg J., Lenk H., DiMichele D., Shapiro A., Tjönnfjord G., Berntorp E. Impact of inhibitor epitope profile on the neutralizing effect against plasma-derived and recombinant factor VIII concentrates in vitro. Haemophilia. 2003;9(5):567–72. doi:10.1046/j.1365-2516.2003.00802.x. PMID: 14511295.; Brackmann H.H., White G.C. 2nd , Berntorp E., Andersen T., Escuriola-Ettingshausen C. Immune tolerance induction: What have we learned over time? Haemophilia. 2018;24 Suppl 3:3–14. doi:10.1111/hae.13445. PMID: 29543371.; Hay C.R., DiMichele D.M.; International Immune Tolerance Study. The principal results of the International Immune Tolerance Study: a randomized dose comparison. Blood. 2012;119(6):1335–44. doi:10.1182/blood-2011-08-369132.; Batsuli G., Zimowski K.L., Tickle K., Meeks S.L., Sidonio R.F. Jr. Immune tolerance induction in paediatric patients with haemophilia A and inhibitors receiving emicizumab prophylaxis. Haemophilia. 2019;25(5):789–96. doi:10.1111/hae.13819.; https://journal.nodgo.org/jour/article/view/858
-
10Academic Journal
المؤلفون: A. V. Poletaev, E. A. Seregina, A. V. Pshonkin, N. A. Karamyan, D. V. Fedorova, S. A. Plyasunova, P. A. Zharkov, А. В. Полетаев, Е. А. Серёгина, А. В. Пшонкин, Н. A. Карамян, Д. В. Фёдорова, С. А. Плясунова, П. А. Жарков
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 8, № 2 (2021); 35-41 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 8, № 2 (2021); 35-41 ; 2413-5496 ; 2311-1267
مصطلحات موضوعية: электрофорез, Willebrand syndrome, factor, diagnosis, multimeric assay, electrophoresis, синдром Виллебранда, фактор, диагностика, мультимерный анализ
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/730/669; Rodeghiero F., Castaman G., Dini E. Epidemiological investigation of the prevalence of von Willebrand's disease. Blood 1987;69:454-9. doi:10.1182/blood.v69.2.454.454.; Nichols W.L., Hultin M.B., James A.H., Manco-Johnson M.J., Montgomery R.R., Ortel T.L., Rick M.E., Sadler J.E., Weinstein M., Yawn B.P. Von Willebrand disease (VWD): evidence-based diagnosis and management guidelines, the National Heart, Lung, and Blood Institute (NHLBI) Expert Panel re- port (USA). Haemophilia 2008;14(2):171-232. doi:10.1111/j.1365-2516.2007.01643.x.; Андреева Т.А., Климова Н.И., Тарасова И.С., Лаврухин Д.Б., Чернов В. М. Состояние специализированной помощи детям и взрослым с болезнью Виллебранда в РФ (по данным анкетирования главных гематологов). Вопросы гематологии/онкологии и иммунопатологии в педиатрии 2013;12(1):5-12.; Leebeek F.W., Eikenboom C.J. Von Willebrand's Disease. N Engl J Med 2016;375(21):2067-80. doi:10.1056/NEJMra1601561.; Yee A., Kretz C.A. Von Willebrand factor: form for function. Semin Thromb Haemost 2014;40(1):17-27. doi:10.1055/s-0033-1363155.; Sadler J.E. A revised classification of von Willebrand disease. For the Subcommittee on von Willebrand Factor of the Scientific and Standardization Committe e of the International Society on Thrombosis and Haemostasis. Thromb Haemost 1994;71(4):520-5. PMID: 8052974.; Sadler J.E., Budde U., Eikenboom J.C.J., Favaloro E.J., Hill F.G.H., Holmberg L., Ingerslev J., Lee C.A., Lillicrap D., Mannucci P.M., Mazurier C., Meyer D., Nichols W.L., Nishino M., Peake I.R., Rodeghiero F., Schneppenheim R., Ruggeri Z.M., Srivastava A., Montgomery R.R., Federici A.B. Update on the pathophysiology and classification of von Willebrand disease: a report of the Subcommittee on von Willebrand Factor. J Thromb Haemost 2006;4:2103-14. doi:10.1111/j.1538-7836.2006.02146.x.; Ledford-Kraemer M.R. Analysis of von Willebrand factor structure by multimer analysis. Am J Hematol 2010;85(7):510-4. doi:10.1002/ajh.21739.; Oliver S., Edwin K., Lau E., Chapman K., Favaloro E. Laboratory Testing for Von Willebrand Factor Multimers. Methods Mol Biol 2017;1646:495-511. doi:10.1007/978-1-4939-7196-1_36.; Collection, Transport, and Processing of Blood Speciments for Testing Plasma-Based Coagulation Assays and Molecular Hemostasis Assays; Approved Guideline-Fifth Edition, 2008. [Electronic resource]. URL: https://clsi.org/media/1399/h21a5_sample.pdf.; Pruthi R.K., Daniels T.M., Heit J.A., Chen D., Owen W.G., Nichols W.L. Plasma von Willebrand factor multimer quantitative analysis by in-gel immunostaining and infrared fluorescent imaging. Thromb Res 2010;126:543-9. doi:10.1016/j.thromres.2010.09.015.; Pikta M., Zemtsovskaja G., Bautista H., Nouadge G., Szanto T., Viigimaa M., Banys V. Preclinical evaluation of a semi-automated and rapid commercial electrophoresis assay for von Willebrand factor multimers. J Clin Lab Anal 2018;32(6):e22416. doi:10.1002/jcla.22416.; Crist R.A., Heikal N.M., Rodgers G.M., Grenache D.G., Smock K.J. Evaluation of a new commercial method for von Willebrand factor multimeric analysis. Int J Lab Hem 2018;40(5):586-91. doi:10.1111/ijlh.12869.; Perez-Rodriguez A., Batlle J., Corrales I., Borras N., Rodriguez-Trillo A., Loures E., Cid A.R., Bonanad S., Cabrera N., Moret A., Parra R., Mingot-Castellano M.E., Navarro N., Altisent C., Perez-Montes R., Marcellini S., Moreto A., Herrero S., Soto I., Mosteirin N.F., Jimenez-Yuste V., Alonso N., de Andres Jacob A., Fontanes E., Campos R., Paloma M.J., Bermejo N., Berrueco R., Mateo J., Arribalzaga K., Marco P., Palomo A., Quismondo N.C., Inigo B., Del Mar Nieto M., Vidal R., Paz Martmez M., Aguinaco R., Tenorio M., Ferreiro M., Garria-Frade J., Rodriguez-Huerta A.M., Cuesta J., Rodriguez-Gonzalez R., Garria-Candel F., Dobon M., Aguilar C., Batlle F., Vidal F., Lopez-Fernandez M.F. Role of multimeric analysis of von Willebrand factor (VWF) in von Willebrand disease (VWD) diagnosis: Lessons from the PCM-EVW-ES Spanish project. PLoS One 2018;13(6):e0197876. doi:10.1371/journal.pone.0197876.; Timm A., Fahrenkrug J., J0rgensen H.L., Sennels H.P., Goetze J.P. Diurnal variation of von Willebrand factor in plasma: the Bispebjerg study of diurnal variations. Eur J Haematol 2014;93(1):48-53. doi:10.1111/ejh.12298.; Bierkaemper S., Trautmann-Grill K., Marten S., Tiebel O., Knoefler R. High variability of laboratory phenotype in genetically proven von Willebrand Disease type 2B. Res Pract Thromb Haemost 2020;4(Suppl 1). ISTH 2020 Virtual Congress.; Полетаев А. В., Серегина Е. А., Федорова Д. В., Жарков П. А. Современные возможности дистанционной диагностики болезни Виллебранда у детей, проживающих на территории Российской Федерации. Вопросы гемотологии/онкологии и иммунопатологии в педиатрии 2020;19(3):54-60. doi:10.24287/1726-1708-2020-19-354-60.; Bykowska K., Ceglarek B. Clinical significance of slightly reduced von Willebrand factor activity. Pol Arch Intern Med 2020;130(3):225-31. doi:10.20452/pamw.15162.; Veyradier A., Jenkins C.S., Fressinaud E., Meyer D. Acquired von Willebrand syndrome: from pathophysiology to management. Tromb Haemost 2000;84:175-82. doi:10.1023/b:thro.0000011369.70824.e6.; https://journal.nodgo.org/jour/article/view/730
-
11Academic Journal
المؤلفون: D. B. Florinskiy, P. A. Zharkov, Д. Б. Флоринский, П. А. Жарков
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 7, № 4 (2020); 71-76 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 7, № 4 (2020); 71-76 ; 2413-5496 ; 2311-1267 ; 10.21682/2311-1267-2020-7-4
مصطلحات موضوعية: профилактика, factor concentrates, cryoprecipitate, fibrinogen, therapy, prevention, концентраты факторов, криопреципитат, фибриноген, терапия
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/670/614; Mannucci P.M., Duga S., Peyvandi F. Recessively inherited coagulation disorders. Blood 2004;104:1243-52. PMID: 15138162.; Mumford A.D., Ackroyd S., Alikhan R., Bowles L., Chowdary P., Grainger J., Mainwaring J., Mathias M., O’Connell N. BCSH Committee Guideline for the diagnosis and management of the rare coagulation disorders: a United Kingdom Haemophilia Centre Doctors' Organization guideline on behalf of the British Committee for Standards in Haematology. Br J Haematol 2014;167:304-26. PMID: 25100430.; Caudill J.S., Nichols W.L., Plumhoff E.A., Schulte S.L., Winters J.L., Gastineau D.A., Rodriguez V. Comparison of coagulation factor XIII content and concentration in cryoprecipitate and fresh-frozen plasma. Transfusion 2009;49(4):765-70. PMID: 19192257.; Зозуля Н. И., Свирин П . В. Диагностика и лечение редких коагулопатий: наследственный дефицит факторов свертывания крови II, VII, X. Национальные клинические рекомендации, 2014 г.; Tabibian S., Yavar S., Shams M., Safa M., Gholami M.S., Heydari F., Ahmadi A., Rashidpanah J., Dorgalaleh A. A comprehensive overview of coagulation factor V and congenital factor V deficiency. Semin Thromb Hemost 2019;45(5):523-43. PMID: 31121608.; Stirling Y., Woolf L., North W.R., Seghatchian M.J., Meade T.W. Haemostasis in normal pregnancy. Thromb Haemost 1984;52:176-82. PMID: 6084322.; Mariani G., Bernardi F. Factor VII deficiency. Semin Thromb Hemost 2009;35(4):400-6. PMID: 20391303.; Woehrle D., Martinez M., Bolliger D. Hereditary heterozygous factor VII deficiency in patients undergoing surgery: clinical relevance. Anaesthesist 2016;65(10):746-54. PMID: 27586406.; Vorstand der Bundesarztekammer auf Empfehlung des Wissenschaftlichen Beirats. Querschnittsleitlinien zur Therapie mit Blutkomponenten und Plasmaderivaten, 2014; 4. uberarbeitete und aktualisierte Auflage.; van Geffen M., Mathijssen N.C.J., Holme P.A., Britta A.P., van Gorkom L., van Kraaij M.G.J., Masereeuw R., Peyvandi F., van Heerde W.L. Pharmacodynamics of recombinant activated factor VII and plasma-derived factor VII in a cohort of severe FVII deficient patients. Thromb Res 2013;132(2):256-62. PMID: 23834817.; Государственный реестр лекарственных средств. Режим доступа: [Электронный ресурс]. URL: http://grls.rosminzdrav.ru/Default.aspx.; Livnat T., Tamarin I., Mor Y., Winckler H., Horowitz Z., Korianski Y., Bar-Zakay B., Seligsohn U., Salomon O. Recombinant activated factor VII and tranexamic acid are haemostatically effective during major surgery in factor XI-deficient patients with inhibitor antibodies. Thromb Haemost 2009;102:487-92. PMID: 19718468.; Dorgalaleh A., Rashidpanah J. Blood coagulation factor XIII and factor XIII deficiency. Blood Rev 2016;30(6):461-75. PMID: 27344554.; Lusher J., Pipe S.W., Alexander S., Nugent D. Prophylactic therapy with Fibrogammin® P is associated with a decreased incidence of bleeding episodes: a retrospective study. Haemophilia 2010;16:316-21. PMID: 20017752.; Karaman S., Akkaya E., Genc S., Bilgili F., Kendirci A.S., Tugcu D., Unuvar A., Karakas Z., Demirkol D., Bayramoglu Z., Omer B. Congenital Factor XIII Deficiency With the Presence of Inhibitor: A Case Study. J Pediatr Hematol Oncol 2019. doi:10.1097/MPH.0000000000001671.; Key N.S., Negrier C. Coagulation factor concentrates: past, present, and future. Lancet 2007;370(9585):439-48. PMID: 17679021.; Spreafico M., Peyvandi F. Combined Factor V and Factor VIII Deficiency. Semin Thromb Hemost 2009;35(4):390-9. PMID: 19598067.; Brenner B., Kuperman A.A., Watzka M., Oldenburg J. Vitamin K-dependent coagulation factors deficiency. Semin Thromb Hemost 2009 Jun;35(4):439-46. PMID: 19598072.; McMahon M.J., James A.H. Combined deficiency of factors II, VII, IX, and X (Borgschulte-Grigsby deficiency) in pregnancy. Obstet Gynecol 2001;97:808-9. PMID: 11336760.; Peyvandi F., Palla R., Menegatti M., Siboni S.M., Halimeh S., Faeser B., Pergantou H., Platokouki H., Giangrande P., Peerlinck K., Celkan T., Ozdemir N., Bidlingmaier C., Ingerslev J., Giansily-Blaizot M., Schved J.F., Gilmore R., Gadisseur A., Benedik-Dolnicar M., Kitanovski L., Mikovic D., Musallam K.M., Rosendaal F.R.; European Network of Rare Bleeding Disorders Group. Coagulation Factor Activity and Clinical Bleeding Severity in Rare Bleeding Disorders: Results From the European Network of Rare Bleeding Disorders J Thromb Haemost 2012;10(4):615-21. PMID: 22321862.; https://journal.nodgo.org/jour/article/view/670
-
12Academic Journal
المؤلفون: D. B. Florinskiy, P. A. Zharkov, Д. Б. Флоринский, П. А. Жарков
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 7, № 3 (2020); 54-63 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 7, № 3 (2020); 54-63 ; 2413-5496 ; 2311-1267 ; 10.21682/2311-1267-2020-7-3
مصطلحات موضوعية: фибриноген, FXII, factor concentrates, cryoprecipitate, fibrinogen, концентраты факторов, криопреципитат
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/625/571; Mannucci P.M., Duga S., Peyvandi F. Recessively inherited coagulation disorders. Blood 2004;104:1243–52. PMID: 15138162.; Palla R., Peyvandi F., Shapiro A.D. Rare bleeding disorders: diagnosis and treatment. Blood 2015;125(13):2052‒61. PMID: 25712993.; de Moerloose P., Casini A., Neerman-Arbez M. Congenital fibrinogen disorders: an update. Semin Thromb Hemost 2013;39:585–95. PMID: 23852822.; Lak M., Keihani M., Elahi F., Peyvandi F., Mannucci P.M. Bleeding and thrombosis in 55 patients with inherited afibrinogenaemia. Br J Haematol 1999;107(1):204–6. PMID: 10520042.; Mackie I.J., Kitchen S., Machin S.J., Lowe G.D. Haemostasis and Thrombosis Task Force of the British Committee for Standards in Haematology. Guidelines on Fibrinogen Assays. Br J Haematol 2003;121(3):396‒404. PMID: 12716361.; Lancellotti S., De Cristofaro R. Congenital prothrombin deficiency. Semin Thromb Hemost 2009;35(4):367‒81. PMID: 19598065.; Tabibian S., Shiravand Y., Shams M., Safa M., Gholami M.S., Heydari F., Ahmadi A., Rashidpanah J., Dorgalaleh A. A comprehensive overview of coagulation factor V and congenital factor V deficiency. Semin Thromb Hemost 2019;45(5):523‒43. PMID: 31121608.; Mariani G., Bernardi F. Factor VII deficiency. Semin Thromb Hemost 2009;35(4):400‒6. PMID: 20391303.; Girolami A., Berti de Marinis G., Bertozzi I., Peroni E., Tasinato V., Lombardi A.M. Discrepant ratios of arterial vs. venous thrombosis in hemophilias A and B as compared to FVII deficiency. Eur J Haematol 2013;91(2):152‒6. PMID: 23621110.; Wojciechowski V.V., Calina D., Tsarouhas K. A guide to acquired vitamin K coagulophathy diagnosis and treatment: the Russian perspective. Daru 2017;25(1):10. PMID: 28416008.; Menegatti M., Peyvandi F. Factor X Deficiency. Semin Thromb Hemost 2009;35(4):407‒15. PMID: 19598069.; Duga S., Salomon O. Factor XI deficiency. Semin Thromb Hemost 2009;35(4):416‒25. PMID: 19598070.; Dorgalaleh A., Rashidpanah J. Blood coagulation factor XIII and factor XIII deficiency. Blood Rev 2016;30(6):461‒75. PMID: 27344554.; Inbal A., Muszbek L. Coagulation factor deficiencies and pregnancy loss. Semin Thromb Hemost 2003;29(2):171–4. PMID: 12709920.; Kohler H., Ichinose A., Seitz R., Ariens R., Muszbek L. Diagnosis and classification of factor XIII deficiencies. J Thromb Haemost 2011;9(7):1404–6. PMID: 22946956.; Spreafico M., Peyvandi F. Combined factor V and factor VIII deficiency. Semin Thromb Hemost 2009;35(4):390‒9. PMID: 19598067.; Brenner B., Kuperman A.A., Watzka M., Oldenburg J. Vitamin K-dependent coagulation factors deficiency. Semin Thromb Hemost 2009;35(4):439‒46. PMID: 19598072.; Boneh A., Bar-Ziv J. Hereditary deficiency of vitamin K dependent coagulation factors with skeletal abnormalities. Am J Med Genet 1996;65:241–3. PMID: 9240751.; Ratnoff O.D., Colopy J.E. A familial hemorrhagic trait associated with a deficiency of a clot‐promoting fraction of plasma. J Clin Invest 1955;34:602–13. PMID: 14367514.; Stavrou E., Schmaier A.H. Factor XII: what does it contribute to our understanding of the physiology and pathophysiology of hemostasis & thrombosis. Thromb Res 2010;125(3):210–5. PMID: 20022081.; Stavrou E.X., Fang C., Bane K.L., Long A.T., Naudin C., Kucukal E., Gandhi A., Brett-Morris A., Mumaw M.M., Izadmehr S., Merkulova A., Reynolds C.C., Alhalabi O., Nayak L., Yu W.-M., Qu C.-K., Meyerson H.J., Dubyak G.R., Gurkan U.A., Nieman M.T., Gupta A.S., Renné T., Schmaier A.H. Factor XII and uPAR upregulate neutrophil functions to influence wound healing. J Clin Invest 2018;128:944–59. PMID: 29376892.; Colman R.W., Schmaier A.H. Contact activation: a vascular biology modulator with anticoagulant, profibrinolytic, antiadhesive, and proinflammatory attributes. Blood 1997;90:3819–43. PMID: 9354649.; Lämmle B., Wuillemin W.A., Huber I., Krauskopf M., Zürcher C., Pflugshaupt R., Furlan M. Thromboembolism and bleeding tendency in congenital factor XII deficiency ‒ a study on 74 subjects from 14 Swiss families. Thromb Haemost 1991;65(2):117‒21. PMID: 1905067.; Yau J.W., Stafford A.R., Liao P., Fredenburgh J.C., Roberts R., Weitz J.I. Mechanism of catheter thrombosis: comparison of the antithrombotic activities of fondaparinux, enoxaparin, and heparin in vitro and in vivo. Blood 2011;118(25):6667–74. PMID: 21937693.; Renné T., Pozgajová M., Grüner S., Schuh K., Pauer H.-U., Burfeind P., Gailani D., Nieswandt B. Defective thrombus formation in mice lacking coagulation factor XII. J Exp Med 2005;202:271–81. PMID: 16009717.; Merkulov S., Zhang W.-M., Komar A.A., Schmaier A.H., Barnes E., Zhou Y., Lu X., Iwaki T., Castellino F.J., Luo G., McCrae K.R. Deletion of murine kininogen gene 1 (mKng1) causes loss of plasma kininogen and delays thrombosis. Blood 2008;111:1274–81. PMID: 18000168.; Revenko A.S., Gao D., Crosby J.R., Bhattacharjee G., Zhao C., May C., Gailani D., Monia B.P., MacLeod A.R. Selective depletion of plasma prekallikrein or coagulation factor XII inhibits thrombosis in mice without increased risk of bleeding. Blood 2011;118(19):5302–11. PMID: 21821705.; Xu Y., Cai T.Q., Castriota G., Zhou Y., Hoos L., Jochnowitz N., Loewrigkeit C., Cook J.A., Wickham A., Metzger J.M., Ogletree M.L., Seiffert D.A., Chen Z. Factor XIIa inhibition by Infestin-4: in vitro mode of action and in vivo antithrombotic benefit. Thromb Haemost 2014;111(4):694‒704. PMID: 24336918.; Peyvandi F., Palla R., Menegatti M., Siboni S.M., Halimeh S., Faeser B., Pergantou H., Platokouki H., Giangrande P., Peerlinck K., Celkan T., Ozdemir N., Bidlingmaier C., Ingerslev J., Giansily-Blaizot M., Schved J.F., Gilmore R., Gadisseur A., Benedik-Dolničar M., Kitanovski L., Mikovic D., Musallam K.M., Rosendaal F.R.; European Network of Rare Bleeding Disorders Group. Coagulation factor activity and clinical bleeding severity in rare bleeding disorders: results from the European Network of Rare Bleeding Disorders. J Thromb Haemost 2012;10(4):615‒21. PMID: 22321862.; https://journal.nodgo.org/jour/article/view/625
-
13Academic Journal
المؤلفون: S. Sh. Uzueva, P. A. Zharkov, С. Ш. Узуева, П. А. Жарков
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 6, № 4 (2019); 69-75 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 6, № 4 (2019); 69-75 ; 2413-5496 ; 2311-1267 ; 10.21682/2311-1267-2019-6-4
مصطلحات موضوعية: дети, venous thrombosis, cancer, solid tumors, children, венозный тромбоз, рак, солидные опухоли
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/556/517; Athale U., Siciliano S., Thabane L., Pai N., Cox S., Lathia A., Khan A., Armstrong A., Chan A.K. Epidemiology and clinical risk factors predisposing to thromboembolism in children with cancer. Pediatr Blood Cancer 2008;51(6):792-7. doi:10.1002/pbc.21734.; Piovesan D., Attard C., Monagle P., Ignjatovic V. Epidemiology of venous thrombosis in children with cancer. Thromb Haemost 2014;111(06):1015–21. doi:10.1160/TH13-10-0827.; Tuckuviene R., Christensen A.L., Helgestad J., Johnsen S.P., Kristensen S.R. Pediatric venous and arterial noncerebral thromboembolism in Denmark: a nationwide population-based study. J Pediatr 2011;159(4):663-9. doi: m.m16/j.jpeds.20n.03.052.; Spencer F., Emery C., Lessard D., Anderson F., Emani S., Aragam J., Becker R.C., Goldberg R.J. The Worcester Venous Thromboembolism study: a population-based study of the clinical epidemiology of venous thromboembolism. J Gen Intern Med 2006;21(7):722-7. doi:10.1111/j.1525-1497.2006.00458.x.; Жарков П.А., Румянцев А.Г, Новичкова ГА. Венозные тромбозы у детей со злокачественными новообразованиями (обзор литературы). Российский журнал детской гематологии и онкологии 2015;2(1):66-74. doi:10.17650/2311-1267-2015-1-66-74.; Raffini L., Huang Y.S., Witmer C., Feudtner C. Dramatic increase in venous thromboembolism in children’s hospitals in the United States from 2001 to 2007. Pediatrics 2009;124(4):1001-8. doi:10.1542/peds.2009-0768.; Walker A.J., Grainge M.J., Card T.R., West J., Ranta S., Ludvigsson J.F. Venous thromboembolism in children with cancer -a population-based cohort study. Thromb Res 2014;133(3):340-4. doi:10.1016/j.thromres.2013.12.021.; Radulescu V., D’Orazio J. Venous Thromboembolic Disease in Children and Adolescents. Adv Exp Med Biol 2017;906:149-65. doi:10.1007/5584_2016_113.; Esmon C. Basic mechanisms and pathogenesis of venous thrombosis. Blood Rev 2009;23(5):225-9. doi:10.1016/j.blre.2009.07.002.; Varki A. Trousseau’s syndrome: multiple definitions and multiple mechanisms. Blood 2007;110:1723-9. doi:10.1182/blood-2006-10-053736.; Carrier M., Le Gal G., Wells P.S., Fergusson D., Ramsay T., Rodger M.A. Systematic review: the Trousseau syndrome revisited: should we screen extensively for cancer in patients with venous thromboembolism? Ann Intern Med 2008;149(5):323-33. doi:10.7326/0003-4819-149-5-200809020-00007.; Skinner R., Koller K., McIntosh N., McCarthy A., Pizer B.; United Kingdom Children’s Cancer Study Group (UKCCSG); Paediatric Oncology Nursing Forum (PONF) Supportive Care Group. Prevention and management of central venous catheter occlusion and thrombosis in children with cancer. Pediatr Blood Cancer 2008;50(4):826-30. doi:10.1002/pbc.21332.; Bajzar L., Chan A., Massicotte M., Mitchell L. Thrombosis in children with malignancy. Curr Opin Pediatr 2006;18(1):1-9. doi:10.1097/01.mop.0000193270.09001.ea.; Petralia G., Lemoine N., Kakkar A. Mechanisms of disease: the impact of antithrombotic therapy in cancer patients. Nat Clin Pract Oncol 2005;2(7):356-63. doi:10.1038/ncponc0225.; Olas B., Wachowicz B., Mielicki W. Cancer procoagulant and blood platelet activation. Cancer Lett 2001;169:165-71. doi:10.1016/s0304-3835(01)00545-6.; Stefansson S., McMahon G., Petitclerc E., Lawrence D. Plasminogen activator inhibitor-1 in tumor growth, angiogenesis and vascular remodeling. Curr Pharm Des 2003;9:1545-64. doi:10.2174/1381612033454621.; Deitcher S., Gajjar A., Kun L., Heideman R.L. Clinically evident venous thromboembolic events in children with brain tumors. J Pediatr 2004;145(6):848-50. doi:10.1016/j.jpeds.2004.05.055.; Athale U. Thrombosis in pediatric cancer: identifying the risk factors to improve care. Exp Rev Hematol 2013;6(5):599-609. doi:10.1586/17474086.2013.842124.; Athale U., Cox S., Siciliano S., Chan A.K. Thromboembolism in children with sarcoma. Pediatr Blood Cancer 2007;49(2):171-6. doi:10.1016/j.thromres.2013.12.021.; Deitcher S. Diagnosis, Treatment, and Prevention of Cancer-Related Venous Thrombosis. Abeloff’s Clinical Oncology (4th ed.), 2008. Part II (46). Pp. 693-711.; Journeycake J.M., Buchanan G.R. Catheter-related deep venous thrombosis and other catheter complications in children with cancer. J Clin Oncol 2006;24(28):4575-80. doi:10.1200/JCO.2009.22.6126.; Kim S., Chung D. Pediatric solid malignancies: neuroblastoma and Wilms’ tumor. Surg Clin North Am 2006;86(2):469-87. doi:10.1016/j.suc.2005.12.008.; Wang J., Chen J.S., Chuang H.Y., Yang Y.J., Chang K.C., Wu J.M. Invasion of the cardiovascular system in childhood malignant hepatic tumors. J Pediatr Hematol Oncol 2002;24(6):436-9. doi:10.1097/00043426-200208000-00006.; Paz-Priel I., Long L., Helman L.J., Mackall C.L., Wayne A.S. Thromboembolic events in children and young adults with pediatric sarcoma. J Clin Oncol 2007;25(12):1519-24. doi:10.1200/JCO.2006.06.9930.; Agnelli G., Bolis G., Capussotti L., Scarpa R.M., Tonelli F., Bonizzoni E., Moia M., Parazzini F., Rossi R., Sonaglia F., Valarani B., Bianchini C., Gussoni G. A clinical outcome-based prospective study on venous thromboembolism after cancer surgery: the @RISTOS project. Ann Surg 2006;243(1):89-95. doi:10.1097/01.sla.0000193959.44677.48.; Vu L., Nobuhara K., Lee H., Farmer D. Determination of risk factors for deep venous thrombosis in hospitalized children. J Pediatr Surg 2008;43:1095-9. doi: m.1016/j.jpedsurg.2008.02.036.; Manco-Johnson M., Beckman M., Goldenberg N. Risk of postthrombotic syndrome development in children with extremity deep venous thrombosis: results of the US centers of disease control and prevention pediatric thrombosis and hemostasis centers. Blood 2009;114:4000. doi: m.n82/blood.V114.22.4000.4000.; Mitchell L., Andrew M., Hanna K., Abshire T., Halton J., Anderson R., Cherrick I., Desai S., Mahoney D., McCuster P., Wu J., Dahl G., Chait P., de Veber G., Lee K.J., Mikulis D., Ginsberg J., Way C.; Prophylactic Antithrombin Replacement in Kids with Acute Lymphoblastic Leukemia Treated with Asparaginase Group (PARKAA). A prospective cohort study determining the prevalence of thrombotic events in children with acute lymphoblastic leukemia and a central venous line who are treated with l-asparaginase: results of the Prophylactic Antithrombin Replacement in Kids with Acute Lymphoblastic Leukemia Treated with Asparaginase (PARKAA) Study. Cancer 2003;97(2):508-16. doi: m.1002/cncr.n042.; Haddad T., Greeno E. Chemotherapy-induced thrombosis. Thromb Res 2006;118:555-8. doi:10.1016/j.thromres.2005.10.015.; Nowak-Gottl U., Wermes C., Junker R., Koch H.G., Schobess R., Fleischhack G., Schwabe D., Ehrenforth S. Prospective evaluation of the thrombotic risk in children with acute lymphoblastic leukemia carrying the MTHFR TT 677 genotype, the prothrombin G20210A variant, and further prothrombotic risk factors. Blood 1999;93:1595-9. doi:10.1182/blood. V93.5.1595.; Forbrigger Z., Kuhle S., Brown M.M., Moorehead P.C., Digout C., Kulkarni K. The association of venous thromboembolism with survival in pediatric cancer patients: a population-based cohort study. Ann Hematol 2018;97(10):1903-8. doi:10.1007/s00277-018-3371-0.; Kuderer N., Culakova E., Lyman G.H., Francis C., Falanga A., Khorana A.A. A validated risk score for venous thromboembolism is predictive of cancer progression and mortality. Oncologist 2016;21(7):861-7. doi:10.1634/theoncologist.2015-0361.; Chew H., Wun T., Harvey D., Zhou H., White R.H. Incidence of venous thromboembolism and its effect on survival among patients with common cancers. Arch Intern Med 2006;166(4):458-64. doi:10.1001/archinte.166.4.458.; Khorana A., Kuderer N.M., Culakova E., Lyman G.H., Francis C.W. Development and validation of a predictive model for chemotherapy-associated thrombosis. Blood 2008;111(10):4902-7. doi:10.1182/blood-2007-10-116327.; Ay C., Dunkler D., Marosi C., Chiriac A.L., Vormittag R., Simanek R., Quehenberger P., Zielinski C., Pabinger I. Prediction of venous thromboembolism in cancer patients. Blood 2010;116(24):5377-82. doi:10.1182/blood-2010-02-270116.; Mitchell L., Lambers M., Flege S., Kenet G., Li-Thiao-Te V., Holzhauer S., Bidlingmaier C., Fruhwald M.C., Heller C., Schmidt W., Pautard B., Nowak-Gottl U. Validation of a predictive model for identifying an increased risk for thromboembolism in children with acute lymphoblastic leukemia: results of a multicenter cohort study. Blood 2010;115(24):4999-5004. doi: 10.U82/blood-2010-01-263012.; Kearon C., Akl E.A., Ornelas J., Blaivas A., Jimenez D., Bounameaux H., Huisman M., King C.S., Morris T.A., Sood N., Stevens S.M., Vintch J.R.E., Wells P., Woller S.C., Moores L. Antithrombotic Therapy for VTE Disease: CHEST Guideline and Expert Panel Report. Chest 2016;149(2):315-52. doi:10.1016/j.chest.2015.11.026.; Lyman G., Bohlke K., Khorana A.A., Kuderer N.M., Lee A.Y., Arcelus J.I., Balaban E.P., Clarke J.M., Flowers C.R., Francis C.W., Gates L.E., Kakkar A.K., Key N.S., Levine M.N., Liebman H.A., Tempero M.A., Wong S.L., Somerfield M.R., Falanga A.; American Society of Clinical Oncology. Venous thromboembolism prophylaxis and treatment in patients with cancer: American society of clinical oncology clinical practice guideline update 2014. J Clin Oncol 2015;33(6):654-6. doi:10.1200/JCO.2014.59.7351.; Farge D., Debourdeau P., Beckers M., Baglin C., Bauersachs R.M., Brenner B., Brilhante D., Falanga A., Gerotzafias G.T., Haim N., Kakkar A.K., Khorana A.A., Lecumberri R., Mandala M., Marty M., Monreal M., Mousa S.A., Noble S., Pabinger I., Prandoni P., Prins M.H., Qari M.H., Streiff M.B., Syrigos K., Bounameaux H., Buller H.R. International clinical practice guidelines for the treatment and prophylaxis of venous thromboembolism in patients with cancer. J Thromb Haemost 2013;11(1):56-70. doi:10.1111/jth.12070.; Prandoni P., Lensing A.W., Piccioli A., Bernardi E., Simioni P., Girolami B., Marchiori A., Sabbion P., Prins M.H., Noventa F., Girolami A. Recurrent venous thromboembolism and bleeding complications during anticoagulant treatment in patients with cancer and venous thrombosis. Blood 2002;100(10):3484-8. doi:10.1182/blood-2002-01-0108.; Monagle P., Chan A.K.C., Goldenberg N.A., Ichord R.N., Journeycake J.M., Nowak-Gottl U., Vesely S.K. Antithrombotic therapy in neonates and children: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012;141(2 Suppl):e737S-e801S. doi:10.1378/chest.11-2308.; Elhasid R., Lanir N., Sharon R., Ben Arush M.W., Levin C., Postovsky S., Ben Barak A., Brenner B. Prophylactic therapy with enoxaparin during L-asparaginase treatment in children with acute lymphoblastic leukemia. Blood Coagul Fibrinolysis 2001;12(5):367-70. doi:10.1097/00001721-200107000-00005.; Harlev D., Zaidman I., Sarig G., Ben Arush M.W., Brenner B., Elhasid R. Prophylactic therapy with enoxaparin in children with acute lymphoblastic leukemia and inherited thrombophilia during L-asparaginase treatment. Thromb Res 2010;126(2):93-7. doi:10.1016/j.thromres.2010.04.013.; Carman T., Kanner A., Barnett G., Deitcher S. Prevention of thromboembolism after neurosurgery for brain and spinal tumors: a survey. South Med J 2003;96:17-22. doi:10.1097/01.SMJ.0000047628.44490.B2.; https://journal.nodgo.org/jour/article/view/556
-
14Academic Journal
المؤلفون: D. A. Gobadze, P. A. Zharkov, Д. А. Гобадзе, П. А. Жарков
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 5, № 1 (2018); 64-67 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 5, № 1 (2018); 64-67 ; 2413-5496 ; 2311-1267 ; 10.17650/2311-1267-2018-5-1
مصطلحات موضوعية: геморрагическая болезнь новорожденных, blood clotting disorders, clotting factors, vitamin K-dependent coagulopathy, intracranial hemorrhage, cholestasis, vitamin K deficiency, hemorrhagic neonatal disease, нарушения свертывания крови, факторы свертывания крови, витамин К-зависимая коагулопатия, внутричерепные кровоизлияния, холестаз, дефицит витамина К
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/360/358; Zhang B., McGee B., Yamaoka J.S. et al. Combined deficiency of factor V and factor VIII is due to mutations in either LMAN1 or MCFD2. Blood 2006;107(5):1903–7. doi:10.1182/blood-2005-09-3620.; Нароган М.В., Карпова А.Л., Строева Л.Е. Витамин К-дефицитный геморрагический синдром у новорожденных и детей первых месяцев жизни. Неонатология: новости, мнения, обучение 2015;3:74–82. [Narogan M.V., Karpova A.L., Stroeva L.E. Vitamin K deficiency bleeding in newborns and children during the first months of life. Neonatologiya: novosti, mneniya, obuchenie = Neonatology: News, Opinions, Training 2015;3:74–82. (In Russ.)].; Ardell S., Offringa M., Soll R. Prophylactic vitamin K for the prevention of vitamin K deficiency bleeding in preterm neonates. Cochrane Database of Systemic Reviews, 2010.; Неонатология. Национальное руководство. Краткое издание. Под ред. акад. РАМН Н.Н. Володина. М.: ГЭОТАР-Медиа, 2013. 896 с. [Neonatology. National leadership. Short edition. Ed. by acad. RAMS N.N. Volodin. M.: GEOTAR-Media, 2013. 896 s. (In Russ.)].; Van Hasselt P.M., de Koning T.J., Kvist N. et al.; Netherlands Study Group for Biliary Atresia Registry. Prevention of vitamin K deficiency bleeding in breastfed infants: Lessons from the Dutch and Danish biliary atresia registries. Pediatrics 2008;121(4):e857–63. doi:10.1542/peds.2007-1788.; Suchy F.J. Neonatal cholestasis. Pediatr Rev 2004;25(11):388–96.; Поляк М.Е., Метелин А.В., Коротеева Н.А. и др. Случай ДНК- диагностики и медико-генетического консультирования прогрессирующего семейного внутрипеченочного холестаза II типа. Клиническая и экспериментальная хирургия. Журнал им. акад. Б.В. Петровского 2015;1:36–41. [Polyak M.E., Metelin A.V., Koroteeva N.A. et al. A case of DNA diagnostic and genetic counseling of progressive familial intrahepatic cholestasis. Klinicheskaya i eksperimentalnaya khirurgiya. Zhurnal im. akad. B.V. Petrovskogo = Clinical and Experimental Surgery. Petrovsky Journal 2015;1:36–41. (In Russ.)].; Hubbard D., Tobias J.D. Intracerebral hemorrhage due to hemorrhagic disease of the newborn and failure to administer vitamin K at birth. South Med J 2006;99(11):1216– 20. doi:10.1097/01.smj.0000233215.43967.69.; American Academy of Pediatrics Committee on Fetus and Newborn. Controversies concerning vitamin K and the newborn. American Academy of Pediatrics Committee on Fetus and Newborn. Pediatrics 2003; 112(1 Pt 1):191–2. PMID: 12837888.; https://journal.nodgo.org/jour/article/view/360
-
15Academic Journal
المؤلفون: N. S. Grachev, S. R. Varfolomeeva, A. I. Karachunskiy, S. R. Talypov, N. A. Bolshakov, I. N. Vorozhtsov, R. S. Oganesyan, M. P. Kalinina, N. V. Babaskina, A. V. Petrushin, A. S. Krasnov, P. D. Pryanikov, P. A. Zharkov, A. G. Rumyantsev, Н. С. Грачёв, С. Р. Варфоломеева, А. И. Карачунский, С. Р. С.Р. Талыпов, Н. А. Большаков, И. Н. Ворожцов, Р. С. Оганесян, М. П. Калинина, Н. В. Бабаскина, А. В. Петрушин, А. С. Краснов, П. Д. Пряников, П. А. Жарков, А. Г. Румянцев
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 3, № 2 (2016); 79-86 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 3, № 2 (2016); 79-86 ; 2413-5496 ; 2311-1267 ; 10.17650/2311-1267-2016-3-2
مصطلحات موضوعية: онкология, orthopedics oncology, endoscopic sinus surgery, microsurgical reconstruction, children, hematology, Oncology, онкоортопедия, эндоскопическая синус-хирургия, микрохирургическая реконструкция, дети, гематология
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/219/215; Simon T., Häberle B., Hero B. et al. Role of surgery in the treatment of patients with stage 4 neuroblastoma age 18 months or older at diagnosis. J Clin Oncol 2013;31(6):752–8.; Buse S., Gilfrich C., Wagener N. et al. Thoracoabdominal approach to large retroperitoneal tumours. BJU Int 2006;98(5):969–72.; Surgue M. Abdominal compartment syndrome. Curr Opin Crit Care 2005;11:333–8.; Hebra A., Powell D.D., Smith C.D. et al. Balloon tracheoplasty in children: results of a 15-year experience. J Pediatr Surg 1991;26:957–61.; Maeda K., Yasuhuku M., Yamamoto T. A new approach to the treatment of congenital tracheal stenosis: balloon tracheoplasty and expandable metallic stenting. J Pediatr Surg 2001;36:1646–9.; Lusk R. Computer-assisted functional endoscopic sinus surgery in children. Otolaryngol Clin North Am 2005;38(3):505–13.; Wise S.K., Del Gaudio J.M. Computer-aided surgery of the paranasal sinuses and skull base. Expert Rev Med Devices 2005;2(4):395–408.; Tschopp K.P., Thomaser E.G. Outcome of functional endonasal sinus surgery with and without CT-navigation. Rhinology 2008;46:116–20.; Liu X.L., Wu C.W., Zhao Y.S. et al. Exclusive real-time monitoring during recurrent laryngeal nerve dissection in conventional monitored thyroidectomy. Kaohsiung J Med Sci 2016;32(3):135–41.; Minahan R.E., Mandir A.S. Neurophysiologic intraoperative monitoring of trigeminal and facial nerves. J Clin Neurophysiol 2011;28(6):551–65.; Ishimaru M., Ono S., Suzuki S. et al. Risk Factors for Free Flap Failure in 2,846 Patients With Head and Neck Cancer: A National Database Study in Japan. J Oral Maxillofac Surg 2016. pii: S0278-2391(16)00024-0. [Epub ahead of print].; Jacob L.M., Dong W., Chang D.W. Outcomes of reconstructive surgery in pediatric oncology patients: Review of 10-year experience. Ann Surg Oncol 2010;17:2563–9.; Rodriguez-Merchan E.C. The haemophilic pseudotumour. Haemophilia 2002;8(1):12–6.; https://journal.nodgo.org/jour/article/view/219