-
1Academic Journal
المؤلفون: T. Z. Aliev, E. B. Machneva, I. O. Kostareva, K. A. Sergeenko, N. A. Burlaka, L. M. Kudaeva, T. I. Potemkina, Yu. V. Lozovan, D. S. Smirnova, A. S. Slinin, K. I. Kirgizov, S. R. Varfolomeeva, Т. З. Алиев, Е. Б. Мачнева, И. О. Костарева, К. А. Сергеенко, Н. А. Бурлака, Л. М. Кудаева, Т. И. Потёмкина, Ю. В. Лозован, Д. С. Смирнова, А. С. Слинин, К. И. Киргизов, С. Р. Варфоломеева
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 10, № 3 (2023); 55-62 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 10, № 3 (2023); 55-62 ; 2413-5496 ; 2311-1267
مصطلحات موضوعية: треосульфан, children, skin, mucositis, complications, hematopoietic stem cell transplantation, toxicity, treosulfan, дети, кожа, мукозит, осложнения, трансплантация гемопоэтических стволовых клеток, токсичность
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/963/847; Алиев Т.З., Мачнева Е.Б., Сидорова Н.В., Белышева Т.С., Валиев Т.Т., Киргизов К.И. Поражение кожных покровов при трансплантации гемопоэтических стволовых клеток. Обзор литературы. Вопросы гематологии/онкологии и иммунопатологии в педиатрии. 2020;19(2):184–92. doi:10.24287/1726-1708-2020-19-2- 184-192.; Белышева Т.С., Алиев Т.З., Валиев Т.Т., Мачнева Е.Б., Сидорова Н.В., Мурашкин Н.Н., Киргизов К.И., Варфоломеева С.Р. Клинические особенности кожной формы острой реакции «трансплантат против хозяина» при аллогенной трансплантации гемопоэтических стволовых клеток у детей с онкогематологическими заболеваниями. Вопросы современной педиатрии. 2020;19(6):500–8. https://doi.org/10.15690/vsp.v19i6.2148.; Bolognia J.L., Cooper D.L., Glusac E.J. Toxic erythema of chemotherapy: a useful clinical term. J Am Acad Dermatol. 2008;59(3):524–9. doi:10.1016/j.jaad.2008.05.018.; Jagasia M.H., Greinix H.T., Arora M., Williams K.M., Wolff D., Cowen E.W., Palmer J., Weisdorf D., Treister N.S., Cheng G.S., Kerr H., Stratton P., Duarte R.F., McDonald G.B., Inamoto Y., Vigorito A., Arai S., Datiles M.B., Jacobsohn D., Heller T., Kitko C.L., Mitchell S.A., Martin P.J., Shulman H., Wu R.S., Cutler C.S., Vogelsang G.B., Lee S.J., Pavletic S.Z., Flowers M.E.D. National Institutes of Health Consensus Development Project on criteria for clinical trials in chronic graft-versus-host disease: I. The 2014 Diagnosis and Staging Working Group report. Biol Blood Marrow Transplant. 2015;21(3):389–401.e1. doi:10.1016/j.bbmt.2014.12.001.; Epstein J.B., Schubert M.M. Oropharyngeal mucositis in cancer therapy. Review of pathogenesis, diagnosis, and management. Oncology (Williston Park). 2003;17(12):1767–79; discussion 1779–82, 1791–2. PMID: 14723014.; Carreras E., Dufour C., Mohty M., Kröger N., eds. The EBMT Handbook: Hematopoietic Stem Cell Transplantation and Cellular Therapies [Internet]. 7th ed. Cham (CH): Springer, 2019. PMID: 32091673.; Wachowiak J., Sykora K.W., Cornish J., Chybicka A., Kowalczyk J.R., Gorczyńska E., Choma M., Grund G., Peters C.; EBMT Pediatric Diseases Working Party. Treosulfan-based preparative regimens for allo-HSCT in childhood hematological malignancies: a retrospective study on behalf of the EBMT pediatric diseases working party. Bone Marrow Transplant. 2011;46(12):1510–8. doi:10.1038/bmt.2010.343.; Główka F.K., Romański M., Wachowiak J. High-dose treosulfan in conditioning prior to hematopoietic stem cell transplantation. Expert Opin Investig Drugs. 2010;19(10):1275–95. doi:10.1517/13543784.2010.517744. PMID: 20836619.; Wachowiak J., Sykora K.W., Cornish J., Chybicka A., Kowalczyk J.R., Gorczynska E., Choma M., Grund G., Peters C. Treosulfan-based preparative regimens for allo-HSCT in childhood hematological malignancies: a retrospective study on behalf of the EBMT Pediatric Diseases Working Party. Bone Marrow Transplant. 2011;46:1510–8. doi:10.1038/bmt.2010.343.; Slatter M.A., Boztug H., Pötschger U., Sykora K.-W., Lankester A., Yaniv I., Sedlacek P., Glogova E., Veys P., Gennery A.R., Peters C., Inborn Errors EBMT, Parties PDW. Treosulfan-based conditioning regimens for allogeneic haematopoietic stem cell transplantation in children with non-malignant diseases. Bone Marrow Transplant. 2015;50:1536–41. doi:10.1038/bmt.2015.171.; Morillo-Gutierrez B., Beier R., Rao K., Burroughs L., Schulz A., Ewins A.M., Gibson B., Sedlacek P., Krol L., Strahm B., Zaidman I., Kalwak K., Talano J.A., Woolfrey A., Fraser C., Meyts I., Müller I., Wachowiak J., Bernardo M.E., Veys P., Sykora K.W., Gennery A.R., Slatter M. Treosulfan-based conditioning for allogeneic HSCT in children with chronic granulomatous disease: a multicenter experience. Blood. 2016;128(3):440–8. doi:10.1182/ blood-2016-03-704015. Erratum in: Blood. 2016;128(21):2585. PMID: 27216217.; Ten Brink M.H., Zwaveling J., Swen J.J., Bredius R.G., Lankester A.C., Guchelaar H.J. Personalized busulfan and treosulfan conditioning for pediatric stem cell transplantation: the role of pharmacogenetics and pharmacokinetics. Drug Discov Today. 2014;19(10):1572–86. doi:10.1016/j.drudis.2014.04.005.; Van der Stoep M.Y.E.C., Bertaina A., Ten Brink M.H., Bredius R.G., Smiers F.J., Wanders D.C.M., Moes D.J.A.R., Locatelli F., Guchelaar H.J., Zwaveling J., Lankester A.C. High interpatient variability of treosulfan exposure is associated with early toxicity in paediatric HSCT: a prospective multicentre study. Br J Haematol. 2017;179(5):772–80. doi:10.1111/bjh.14960.; Kearns G.L., Abdel-Rahman S.M., Alander S.W., Blowey D.L., Leeder J.S., Kauff man R.E. Developmental pharmacology–drug disposition, action, and therapy in infants and children. N Engl J Med. 2003;349(12):1157–67. doi:10.1056/NEJMra035092.; Scheulen M.E., Hilger R.A., Oberhoff C., Casper J., Freund M., Josten K.M., Bornhäuser M., Ehninger G., Berdel W.E., Baumgart J., Harstrick A., Bojko P., Wolf H.H., Schindler A.E., Seeber S. Clinical phase I dose escalation and pharmacokinetic study of high-dose chemotherapy with treosulfan and autologous peripheral blood stem cell transplantation in patients with advanced malignancies. Clin Cancer Res. 2000;6(11):4209–16. PMID: 11106234.; Rosman I.S., Lloyd B.M., Hayashi R.J, Bayliss S.J. Cutaneous effects of thiotepa in pediatric patients receiving high-dose chemotherapy with autologous stem cell transplantation. J Am Acad Dermatol. 2008;58(4):575–8. doi:10.1016/j.jaad.2007.12.037.; Tkachenko P.E., Ivashkin V.T., Maevskaya M.V. Clinical guidelines for the correction of hepatotoxicity induced by anticancer therapy. Malignant tumors. 2020;10(3s2-2):52–65. doi:10.18027/2224-5057-2020-10-3s2-40.; Иваников И.О., Виноградова Н.Н., Крашенков О.П., Коцелапова Э.Ю., Григорьева Ю.В., Мясникова Е.В., Пикунова И.Н. Острые повреждения печени вследствие химиотерапии и возможности их лечения. Доказательная гастроэнтерология. 2020;9(4):7–15. doi:10.17116/dokgastro202090417.; Levi M., Stemmer S.M., Stein J., Shalgi R., Ben-Aharon I. Treosulfan induces distinctive gonadal toxicity compared with busulfan. Oncotarget. 2018;9(27):19317–27. doi:10.18632/oncotarget.25029.; Feit P.W., Rastrup-Andersen N., Matagne R. Studies on epoxide formation from (2S,3S)-threitol 1,4-bismethanesulfonate. The preparation and biological activity of (2S,3S)-1,2-epoxy-3,4- butanediol 4-methanesulfonate. J Med Chem. 1970;13(6):1173–5. doi:10.1021/jm00300a034. PMID: 5479859.; Romański M., Ratajczak W., Główka F. Kinetic and mechanistic study of the pH-dependent activation (epoxidation) of prodrug treosulfan including the reaction inhibition in a borate buff er. J Pharm Sci. 2017;106:1917–22. doi:10.1016/j.xphs.2017.03.018.; https://journal.nodgo.org/jour/article/view/963
-
2Academic Journal
المؤلفون: A. D. Shirin, R. Ya. Vlasenko, N. Yu. Anisimova, K. I. Kirgizov, T. T. Valiev, N. G. Stepanyan, T. Z. Aliev, G. E. Morozevich, O. A. Odaryuk, D. V. Filonenko, N. E. Nifantiev, K. M. Novruzov, I. O. Chikileva, M. V. Kiselevskiy, А. Д. Ширин, Р. Я. Власенко, Н. Ю. Анисимова, К. И. Киргизов, Т. Т. Валиев, Н. Г. Степанян, Т. З. Алиев, Г. Е. Морозевич, О. А. Одарюк, Д. В. Филоненко, Н. Э. Нифантьев, К. М. Новрузов, И. О. Чикилева, М. В. Киселевский
المساهمون: This work was supported by the Russian Foundation for Basic Research (RFBR-comfi grant: projects nos. 17-00-00494, 17-00-00495 and 17-00-00496), Работа выполнена при финансовой поддержке Российского фонда фундаментальных исследований (грант РФФИ- комфи: проекты № 17-00-00494, 17-00-00495 и 17-00-00496)
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 9, № 4 (2022); 64-74 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 9, № 4 (2022); 64-74 ; 2413-5496 ; 2311-1267
مصطلحات موضوعية: экстренный гемопоэз, hematopoiesis, Toll-like receptor inductors, emergency hematopoiesis, гемопоэз, индукторы Toll-подобных рецепторов
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/884/781; Passweg J. R., Baldomero H., Bader P., Bonini C., Cesaro S., Dreger P., Duarte R. F., Dufour C., Kuball J., Farge-Bancel D., Gennery A., Kröger N., Lanza F., Nagler A., Sureda A., Mohty M. Hematopoietic stem cell transplantation in Europe 2014: More than 40 000 transplants annually. Bone Marrow Transplant. 2016; 51 (6): 786–92. doi:10.1038/bmt.2016.20.; Lee K. H., Lee J. H., Choi S. J. Failure of trilineage blood cell reconstitution after initial neutrophil engraftment in patients undergoing allogeneic hematopoietic cell transplantation–frequency and outcomes. Bone Marrow Transplant. 2004; 33 (7): 729–34. doi:10.1038/sj.bmt.1704428.; Lin Y., Hu X., Cheng H. Graft-versus-host disease causes broad suppression of hematopoietic primitive cells and blocks megakaryocyte diff erentiation in a murine model. Biol Blood Marrow Transplant. 2014; 20 (9): 1290–300. doi:10.1016/j.bbmt.2014.05.009.; Kuzmina Z., Eder S., Bohm A. Signifi cantly worse survival of patients with NIH-defi ned chronic graft-versus-host disease and thrombocytopenia or progressive onset type: Results of a prospective study. Leukemia. 2011; 26 (4): 746–56. doi:10.1038/leu.2011.257.; Müskens K. F., Lindemans C. A., Belderbos M. E. Hematopoietic Dysfunction during Graft-Versus-Host Disease: A Self-Destructive Process? Cells. 2021; 10 (8): 2051. doi:10.3390/cells10082051.; Ferrara J. L., Levine J. E., Reddy P., Holler E. Graft-versus-host disease. Lancet. 2009; 373 (9674): 1550–61. doi:10.1016/S0140-6736(09)60237-3.; Danziger-Isakov L., Baillie M. G. Hematologic complications of anti-CMV therapy in solid organ transplant recipients. Clin Transplant. 2009; 23 (3): 295–304. doi:10.1111/j.1399-0012.2008.00942.x.; Andersohn F., Konzen C., Garbe E. Systematic review: Agranulocytosis induced by nonchemotherapy drugs. Ann Intern Med. 2007; 146 (9): 657–65. doi:10.7326/0003-4819-146-9-200705010-00009.; Rizzo J. D., Somerfield M. R., Hagerty K. L., Seidenfeld J., Bohlius J., Bennett C. L., Cella D. F., Djulbegovic B., Goode M. J., Jakubowski A. A., Rarick M. U., Regan D. H., Lichtin A. E. Use of epoetin and darbepoetin in patients with cancer: 2007 American Society of Hematology / American Society of Clinical Oncology clinical practice guideline update. Blood. 2008; 111 (1): 25–41. doi:10.1182/blood-2007-08-109488.; Bokemeyer C., Aapro M. S., Courdi A., Foubert J., Link H., Osterborg A., Repetto L., Soubeyran P. European Organisation for Research and Treatment of Cancer (EORTC) Taskforce for the Elderly. EORTC guidelines for the use of erythropoietic proteins in anaemic patients with cancer: 2006 update. Eur J Cancer. 2007; 43 (2): 258–70. doi:10.1016/j.ejca.2006.10.014.; Ivanov V., Faucher C., Mohty M., Bilger K., Ladaique P., Sainty D., Arnoulet C., Chabannon C., Vey N., Camerlo J., Bouabdallah R., Maraninchi D., Bardou V. J., Blaise D. Decreased RBCTs after reduced intensity conditioning allogeneic stem cell transplantation: predictive value of prior Hb level. Transfusion. 2004; 44 (4): 501–8. doi:10.1111/j.1537-2995.2004.03317.x.; NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®). Myelodysplastic Syndromes Version 3.2022 – January 13, 2022. [Electronic resource]: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1446 (appeal date 02. 07. 2022).; Beguin Y., Baron F., Fillet G. Influence of marrow erythropoietic activity on serum erythropoietin levels after autologous hematopoietic stem cell transplantation. Haematologica. 1998; 83 (12): 1076–81. PMID: 9949624.; Pene R., Appelbaum F. R., Fisher L., Lilleby K., Nemunaitis J., Storb R., Buckner C. D. Use of granulocyte-macrophage colony-stimulating factor and erythropoietin in combination after autologous marrow transplantation. Bone Marrow Transplant. 1993; 11 (3): 219–22. PMID: 8467286.; Vannucchi A. M., Bosi A., Ieri A., Guidi S., Saccardi R., Lombardini L., Linari S., Laszlo D., Longo G., Rossi-Ferrini P. Combination therapy with G-CSF and erythropoietin after autologous bone marrow transplantation for lymphoid malignancies: A randomized trial. Bone Marrow Transplant. 1996; 17 (4): 527–31. PMID: 8722349.; Baron F., Frere P., Fillet G., Beguin Y. Recombinant human erythropoietin therapy is very effective after an autologous peripheral blood stem cell transplant when started soon after engraftment. Clin Cancer Res. 2003; 9 (15): 5566–72. PMID: 14654537.; Beguin Y., Maertens J., De Prijck B., Schots R., Seidel L., Bonnet C., Hafraoui K., Willems E., Vanstraelen G., Servais S., Jaspers A., Fillet G., Baron F. Darbepoetin-alfa and intravenous iron administration after autologous hematopoietic stem cell transplantation: A prospective multicenter randomized trial. Am J Hematol. 2013; 88 (12): 990–6. doi:10.1002/ajh.23552.; Bohlius J., Schmidlin K., Brillant C., Schwarzer G., Trelle S., Seidenfeld J., Zwahlen M., Clarke M., Weingart O., Kluge S., Piper M., Rades D., Steensma D. P., Djulbegovic B., Fey M. F., Ray-Coquard I., Machtay M., Moebus V., Thomas G., Untch M., Schumacher M., Egger M., Engert A. Recombinant human erythropoiesis-stimulating agents and mortality in patients with cancer: A meta-analysis of randomised trials. Lancet. 2009; 373 (9674): 1532–42. doi:10.1016/s0140-6736(09)60502-x.; Bennett C. L., Silver S. M., Djulbegovic B., Samaras A. T., Blau C. A., Gleason K. J., Barnato S. E., Elverman K. M., Courtney D. M., McKoy J. M., Edwards B. J., Tigue C. C., Raisch D. W., Yarnold P. R., Dorr D. A., Kuzel T. M., Tallman M. S., Trifilio S. M., West D. P., Lai S. Y., Henke M. Venous thromboembolism and mortality associated with recombinant erythropoietin and darbepoetin administration for the treatment of cancer-associated anemia. JAMA. 2008; 299 (8): 914–24. doi:10.1001/jama.299.8.914.; Demetri G. D., Kris M., Wade J., Degos L., Cella D. Quality-of-life benefit in chemotherapy patients treated with epoetin alfa is independent of disease response or tumor type: Results from a prospective community oncology study. Procrit Study Group. J Clin Oncol. 1998; 16 (10): 3412–25. doi:10.1200/jco.1998.16.10.3412.; Taher A. T., Musallam K. M., Cappellini M. D. β-Thalassemias. N Engl J Med. 2021; 384 (8): 727–43. doi:10.1056/NEJMra2021838.; Singbart G. Adverse events of erythropoietin in long-term and in acute / short-term treatment. Clin Investig. 1994; 72 (6 Suppl): S36–43. PMID: 7950171.; Снеговой А. В. Практические рекомендации по назначению колониестимулирующих факторов с целью профилактики развития фебрильной нейтропении у онкологических больных / А. В. Снеговой [и др.] // Злокачественные опухоли. – 2015. –4 (S): 342–9. URL: https://rosoncoweb.ru/standarts/RUSSCO/2016/35.pdf.; Ernst P., Bacigalupo A., Ringdén O., Ruutu T., Kolb H. J., Lawrinson S., Skacel T. A phase 3, randomized, placebo-controlled trial of filgrastim in patients with haematological malignancies undergoing matched-related allogeneic bone marrow transplantation. Arch Drug Inf. 2008; 1 (3): 89–96. doi:10.1111/j.1753-5174.2008.00013.x.; Bishop M. R., Tarantolo S. R., Geller R. B., Lynch J. C., Bierman P. J., Pavletic Z. S., Vose J. M., Kruse S., Dix S. P., Morris M. E., Armitage J. O., Kessinger A. A randomized, double-blind trial of fi lgrastim (granulocyte colony-stimulating factor) versus placebo following allogeneic blood stem cell transplantation. Blood. 2000; 96 (1): 80–5. doi:10.1182/blood.v96.1.80.013k35_80_85.; Ringden O. T., Le Blanc K., Remberger M. Granulocyte and granulocyte-macrophage colony-stimulating factors in allografts: Uses, misuses, misconceptions, and future applications. Exp Hematol. 2005; 33 (5): 505–12. doi:10.1016/j.exphem.2005.01.009.; Update of recommendations for the use of hematopoietic colony-stimulating factors: Evidence-based clinical practice guidelines. American Society of Clinical Oncology J Clin Oncol. 1996; 14 (4): 1957–60. doi:10.1200/jco.1996.14.6.1957.; Khoury H. J., Loberiza F. R. Jr., Ringdén O., Barrett A. J., Bolwell B. J., Cahn J. Y., Champlin R. E., Gale R. P., Hale G. A., Urbano-Ispizua A., Martino R., McCarthy P. L., Tiberghien P., Verdonck L. F., Horowitz M. M. Impact of posttransplantation G-CSF on outcomes of allogeneic hematopoietic stem cell transplantation. Blood. 2006; 107 (4): 1712–6. doi:10.1182/blood-2005-07-2661.; Ringden O., Labopin M., Gorin N. C. Treatment with granulocyte colony-stimulating factor after allogeneic bone marrow transplantation for acute leukemia increases the risk of graft-versus-host disease and death: A study from the Acute Leukemia Working Party of the European Group for Blood and Marrow Transplantation J Clin Oncol. 2004; 22 (3): 416–23. doi:10.1200/jco.2004.06.102.; Morris E. S., MacDonald K. P., Kuns R. D., Morris H. M., Banovic T., Don A. L., Rowe V., Wilson Y. A., Raffelt N. C., Engwerda C. R., Burman A. C., Markey K. A., Godfrey D. I., Smyth M. J., Hill G. R. Induction of natural killer T cell-dependent alloreactivity by administration of granulocyte colony-stimulating factor after bone marrow transplantation Nat Med. 2009; 15 (4): 436–41. doi:10.1038/nm.1948.; Kim S., Baek J., Min H. Effects of prophylactic hematopoietic colony stimulating factors on stem cell transplantations: meta-analysis. Arch Pharm Res. 2012; 35 (11): 2013–20. doi:10.1007/s12272-012-1119-2.; Ringden O., Hassan Z., Karlsson H., Olsson R., Omazic B., Mattsson J., Remberger M. Granulocyte colony-stimulating factor induced acute and chronic graft-versus-host disease. Transplantation. 2010; 90 (9): 1022–9. doi:10.1097/TP.0b013e3181f585c7.; Büchner T. Hematopoietic growth factors in cancer treatment. Stem Cells. 1994; 12 (3): 241–52. doi:10.1002/stem.5530120301.; Karagiannidis I., Salataj E., Said Abu Egal E., Beswick E. J. G-CSF in tumors: aggressiveness, tumor microenvironment and immune cell regulation. Cytokine. 2021; 142: 155479. doi:10.1016/j.cyto.2021.155479.; Rodeghiero F. A critical appraisal of the evidence for the role of splenectomy in adults and children with ITP. Br J Haematol. 2018; 181 (2): 183–95. doi:10.1111/bjh.15090.; Ghanima W., Godeau B., Cines D. B., Bussel J. B. How I treat immune thrombocytopenia: the choice between splenectomy or a medical therapy as a second-line treatment. Blood. 2012; 120 (5): 960–9. doi:10.1182/blood-2011-12-309153.; Will B., Kawahara M., Luciano J. P., Bruns I., Parekh S., Erickson-Miller C. L., Aivado M. A., Verma A., Steidl U. Effect of the nonpeptide thrombopoietin receptor agonist Eltrombopag on bone marrow cells from patients with acute myeloid leukemia and myelodysplastic syndrome. Blood. 2009; 114 (18): 3899–908. doi:10.1182/blood-2009-04-219493.; Di Buduo C. A., Currao M., Pecci A., Kaplan D. L., Balduini C. L., Balduini A. Revealing eltrombopag’s promotion of human megakaryopoiesis through AKT / ERK-dependent pathway activation. Haematologica. 2016; 101 (12): 1479–88. doi:10.3324/haematol.2016.146746.; Bussel J. B., Buchanan G. R., Nugent D. J. A randomized, double-blind study of romiplostim to determine its safety and effi cacy in children with immune thrombocytopenia. Blood. 2011; 118 (1): 28–36. doi:10.1182/blood-2010-10-313908.; Wang L., Gao Z., Chen X. P., Zhang H. Y., Yang N., Wang F. Y., Guan L. X., Gu Z. Y., Zhao S. S., Luo L., Wei H. P., Gao C. J. Efficacy and safety of thrombopoietin receptor agonists in patients with primary immune thrombocytopenia: A systematic review and meta-analysis. Sci Rep. 2016; 6: 39003. doi:10.1038/srep39003.; Kuzmina Z., Eder S., Böhm A., Pernicka E., Vormittag L., Kalhs P., Petkov V., Stary G., Nepp J., Knobler R., Just U., Krenn K., Worel N., Greinix H. T. Signifi cantly worse survival of patients with NIH-defi ned chronic graft-versus-host disease and thrombocytopenia or progressive onset type: Results of a prospective study. Leukemia. 2012; 26 (4): 746–56. doi:10.1038/leu.2011.257.; Kim D. H., Sohn S. K., Baek J. H., Kim J. G., Lee N. Y., Won D. I., Suh J. S., Lee K. B. Clinical signifi cance of platelet count at day +60 after allogeneic peripheral blood stem cell transplantation. J Korean Med Sci. 2006; 21 (1): 46–51. doi:10.3346/jkms.2006.21.1.46.; Diedrich B., Remberger M., Shanwell A., Svahn B. M., Ringdén O. A prospective randomized trial of a prophylactic platelet transfusion trigger of 10 × 109 per L versus 30 × 109 per L in allogeneic hematopoietic progenitor cell transplant recipients. Transfusion. 2005; 459 (7): 1064–72. doi:10.1111/j.1537-2995.2005.04157.x.; Liu X., Wu M., Peng Y., Chen X., Sun J., Huang F., Fan Z., Zhou H., Wu X., Yu G., Zhang X., Li Y., Xiao Y., Song C., Xiang A. P., Liu Q. Improvement in poor graft function after allogeneic hematopoietic stem cell transplantation upon administration of mesenchymal stem cells from third-party donors: A pilot prospective study. Cell Transplant. 2014; 23 (9): 1087–98. doi:10.3727/096368912X661319.; Ahmed S., Bashir Q., Bassett R., Poon M. C., Valdez B., Konoplev S., Alousi A. M., Andersson B. S., Ciurea S., Hosing C., Jones R., Kebriaei P., Khouri I., Kim S., Nieto Y., Olson A., Oran B., Parmar S., Qazilbash M. H., Rezvani K., Shah N., Shpall E. J., Champlin R., Popat U. Eltrombopag for Post-Transplantation Thrombocytopenia: Results of Phase II Randomized, Double-Blind, Placebo-Controlled Trial. Transplant Cell Ther. 2021; 27 (5): 430.e1–430.e7. doi:10.1016/j.jtct.2021.02.004.; Kim T. O., Despotovic J., Lambert M. P. Eltrombopag for use in children with immune thrombocytopenia. Blood Adv. 2018; 2 (4): 454–61. doi:10.1182/bloodadvances.2017010660.; Gonzalez-Porras J. R., Bastida J. M. Eltrombopag in immune thrombocytopenia: Efficacy review and update on drug safety. Ther Adv Drug Saf. 2018; 9 (6): 263–85. doi:10.1177/2042098618769587.; McHutchison J. G., Dusheiko G., Shiffman M. L., Rodriguez-Torres M., Sigal S., Bourliere M., Berg T., Gordon S. C., Campbell F. M., Theodore D., Blackman N., Jenkins J., Afdhal N. H. TPL102357 Study Group. Eltrombopag for thrombocytopenia in patients with cirrhosis associated with hepatitis C. N Engl J Med. 2007; 357 (22): 2227–36. doi:10.1056/NEJMoa073255.; Marsh J. C. W., Mufti G. J. Eltrombopag: A stem cell cookie? Blood. 2014; 123 (12): 1774–5. doi:10.1182/blood-2014-02-553404.; Erickson-Miller C. L., Delorme E., Tian S. S., Hopson C. B., Landis A. J., Valoret E. I., Sellers T. S., Rosen J., Miller S. G., Luengo J. I., Duffy K. J., Jenkins J. M. Preclinical Activity of Eltrombopag (SB-497115), an Oral, Nonpeptide Thrombopoietin Receptor Agonist. Stem Cells. 2009; 27 (2): 424–30. doi:10.1634/stemcells.2008-0366.; Alvarado L. J., Huntsman H. D., Cheng H., Townsley D. M., Winkler T., Feng X., Dunbar C. E., Young N. S., Larochelle A. Eltrombopag maintains human hematopoietic stem and progenitor cells under inflammatory conditions mediated by IFNγ. Blood. 2019; 133 (19): 2043–55. doi:10.1182/blood-2018-11-884486.; Yaman Y., Elli M., Şahin Ş., Özdilli K., Bilgen H., Bayram N., Nepesov S., Anak S. Eltrombopag for treatment of thrombocytopenia after allogeneic hematopoietic cell transplantation in children: Single-centre experience. Pediatr Transplant. 2021; 25 (5): e13962. doi:10.1111/petr.13962.; Fu H., Zhang X., Han T., Mo X., Wang Y., Chen H., Han W., Wang J., Wang F., Yan C., Zhang Y., Sun Y., Liu K., Huang X., Xu L. Eltrombopag is an effective and safe therapy for refractory thrombocytopenia after haploidentical hematopoietic stem cell transplantation. Bone Marrow Transplant. 2019; 54 (8): 1310–8. doi:10.1038/s41409-019-0435-2.; Bento L., Bastida J. M., García-Cadenas I., García-Torres E., Rivera D., Bosch-Vilaseca A., De Miguel C., Martínez-Muñoz M. E., Fernández-Avilés F., Roldán E., Chinea A., Yáñez L., Zudaire T., Vaz C. P., Espigado I., López J., Valcárcel D., Duarte R., Cabrera R., Herrera C., González-Porras J. R., Gutiérrez A., Solano C., Sampol A. Grupo Español de Trasplante Hematopoyético (GETH). Thrombopoietin Receptor Agonists for Severe Thrombocytopenia after Allogeneic Stem Cell Transplantation: Experience of the Spanish Group of Hematopoietic Stem Cell Transplant. Biol Blood Marrow Transplant. 2019; 25 (9): 1825–31. doi:10.1016/j.bbmt.2019.05.023.; Mittelman M., Platzbecker U., Afanasyev B., Grosicki S., Wong R. S. M., Anagnostopoulos A., Brenner B., Denzlinger C., Rossi G., Nagler A., Garcia-Delgado R., Portella M. S. O., Zhu Z., Selleslag D. Eltrombopag for advanced myelodysplastic syndromes or acute myeloid leukaemia and severe thrombocytopenia (ASPIRE): A randomised, placebo-controlled, phase 2 trial. Lancet Haematol. 2018; 5 (1): e34–e43. doi:10.1016/S2352-3026(17)30228-4.; Christakopoulos G. E., DeFor T. E., Hage S., Wagner J. E., Linden M. A., Brunstein C., Bejanyan N., Verneris M. R., Smith A. R. Phase I dose-finding, safety and tolerability trial of Romiplostim to Improve Platelet Recovery after UCB Transplantation. Transplant Cell Ther. 2021; 27 (6): 497.e1–497.e6. doi:10.1016/j.jtct.2021.02.033.; Calmettes C., Vigouroux S., Tabrizi R., Milpied N. Romiplostim (AMG531, Nplate) for secondary failure of platelet recovery after allo-SCT. Bone Marrow Transplant. 2011; 46 (12): 1587–9. doi:10.1038/bmt.2011.179.; Battipaglia G., Ruggeri A., Brissot E., Mamez A. C., Malard F., Belhocine R., Vekhoff A., Giannotti F., Ledraa T., Labopin M., Rubio M. T., Mohty M. Safety and feasibility of romiplostim treatment for patients with persistent thrombocytopenia after allogeneic stem cell transplantation. Bone Marrow Transplant. 2015; 50 (12): 1574–7. doi:10.1038/bmt.2015.182.; Maximova N., Zanon D., Rovere F., Maestro A., Schillani G., Paparazzo R. Romiplostim for secondary thrombocytopenia following allogeneic stem cell transplantation in children. Int J Hematol. 2015; 102 (5): 626–32. doi:10.1007/s12185-015-1821-1.; Hartranft M. E., Clemmons A. B., Deremer D. L., Kota V. Evaluation of romiplostim for the treatment of secondary failure of platelet recovery among allogeneic hematopoietic stem cell transplant patients. J Oncol Pharm Pract. 2017; 23 (1): 10–7. doi:10.1177/1078155215612240.; Kantarjian H., Fenaux P., Sekeres M. A. Becker P. S., Boruchov A., Bowen D., Hellstrom-Lindberg E., Larson R. A., Lyons R. M., Muus P., Shammo J., Siegel R., Hu K., Franklin J., Berger D. P. Safety and efficacy of romiplostim in patients with lower-risk myelodysplastic syndrome and thrombocytopenia. J Clin Oncol. 2010; 28 (3): 437–44. doi:10.1200/JCO.2009.24.7999.; Sekeres M. A., Kantarjian H., Fenaux P., Becker P., Boruchov A., Guerci-Bresler A., Hu K., Franklin J., Wang Y. M., Berger D. Subcutaneous or intravenous administration of romiplostim in thrombocytopenic patients with lower risk myelodysplastic syndromes. Cancer. 2011; 117 (5): 992–1000. doi:10.1002/cncr.25545.; Grainger J. D., Locatelli F., Chotsampancharoen T. Eltrombopag for children with chronic immune thrombocytopenia (PETIT2): a randomised, multicentre, placebo-controlled trial. Lancet. 2015; 386 (10004): 1649–58. doi:10.1016/s0140-6736(15)61107-2.; Cuker A., Chiang E. Y., Cines D. B. Safety of the thrombopoiesis-stimulating agents for the treatment of immune thrombocytopenia. Curr Drug Saf. 2010; 5 (2): 171–81. URL: https://pubmed.ncbi.nlm.nih.gov/19534637/.; Bento L., Canaro M., Bastida J. M., Sampol A. Thrombocytopenia and Therapeutic Strategies after Allogeneic Hematopoietic Stem Cell Transplantation. J Clin Med. 2022; 11 (5): 1364. doi:10.3390/jcm11051364.; Luo S. S., Ogata K., Yokose N., Kato T. Dan K. Effect of thrombopoietin on proliferation of blasts from patients with myelodysplastic syndromes. Stem Cells. 2000; 18 (2): 112–9. doi:10.1634/stemcells.18-2-112.; Hashimoto S., Toba K., Fuse I. Thrombopoietin activates the growth of megakaryoblasts in patients with chronic myeloproliferative disorders and myelodysplastic syndrome. Eur J Haematol. 2000; 64 (4): 225–30. doi:10.1034/j.1600-0609.2000.90001.x.; O’Driscoll D. N. Emergency myelopoiesis in critical illness: lessons from the COVID-19 pandemic. Ir J Med Sci. 2022; 16: 1–2. doi:10.1007/s11845-022-03068-w.; Mitroulis I., Kalafati L., Hajishengallis G., Chavakis T. Myelopoiesis in the Context of Innate Immunity. J Innate Immun. 2018; 10 (5–6): 365–72. doi:10.1159/000489406.; Trumpp A., Essers M., Wilson A. Awakening dormant haematopoietic stem cells. Nat Rev Immunol. 2010; 10 (3): 201–9. doi:10.1038/nri2726.; Kuderer N. M., Dale D. C., Crawford J., Cosler L. E., Lyman G. H. Mortality, morbidity, and cost associated with febrile neutropenia in adult cancer patients. Cancer. 2006; 106 (10): 2258–66. doi:10.1002/cncr.21847.; Hérault A., Binnewies M., Leong S., Calero-Nieto F. J., Zhang S. Y., Kang Y. A., Wang X., Pietras E. M., Chu S. H., Barry-Holson K., Armstrong S., Göttgens B., Passegué E. Myeloid progenitor cluster formation drives emergency and leukaemic myelopoiesis. Nature. 2017; 544 (7648): 53–8. doi:10.1038/nature21693.; Nagai Y., Garrett K. P., Ohta S., Bahrun U., Kouro T., Akira S., Takatsu K., Kincade P. W. Toll-like receptors on hematopoietic progenitor cells stimulate innate immune system replenishment. Immunity. 2006; 24 (6): 801–12. doi:10.1016/j.immuni.2006.04.008.; Boettcher S., Ziegler P., Schmid M. A., Takizawa H., van Rooijen N., Kopf M., Heikenwalder M., Manz M. G. Cutting Edge: LPS-Induced Emergency Myelopoiesis Depends on TLR4-Expressing Nonhematopoietic Cells. J Immunol. 2012; 188 (12): 5824–8. doi:10.4049/jimmunol.1103253.; Калюжин О. В. Мурамилпептиды в эксперименте и клинике / О. В. Калюжин // Журнал микробиологии эпидемиологии и иммунобиологии. – 1998. – 1: 104–8.; Львов В. Л. Применение композиции, состоящей из низкомолекулярных фрагментов пептидогликана грамотрицательных бактерий, для лечения и профилактики заболеваний человека / В. Л. Львов [и др.] // Патент РФ. – RU2441906C2. – 2012.; Маркова Т. П. Мурамилпептиды: механизмы действия, клиническая эффективность и перспективы применения в медицине / Т. П. Маркова [и др.] // РМЖ. Медицинское обозрение. – 2020. – 4 (1): 31–7. doi:10.32364/2587-6821-2020-4-1-31-37.; Ustyuzhanina N. E., Anisimova N. Y., Bilan M. I., Donenko F. V., Morozevich G. E., Yashunskiy D. V., Usov A. I., Siminyan N. G., Kirgisov K. I., Varfolomeeva S. R., Kiselevskiy M. V., Nifantiev N. E. Chondroitin Sulfate and Fucosylated Chondroitin Sulfate as Stimulators of Hematopoiesis in Cyclophosphamide-Induced Mice. Pharmaceuticals (Basel). 2021; 14 (11): 1074. doi:10.3390/ph14111074.; Киселевский М. В. Комбинация мурамилпептидов грамотрицательных бактерий корригирует нарушения гемопоэза и клеточного состава селезенки, вызванные циклофосфамидом, у мышей с меланомой B16 / М. В. Киселевский [и др.] // Бюллетень экспериментальной биологии и медицины. – 2020. –170 (12): 772–7. doi:10.47056/0365-9615-2020-170-12-772-777.; Hsu H. Y., Lin T. Y., Lu M. K., Leng P. J., Tsao S. M., Wu Y. C. Fucoidan induces Toll-like receptor 4-regulated reactive oxygen species and promotes endoplasmic reticulum stress-mediated apoptosis in lung cancer. Sci Rep. 2017; 7: 44990. doi:10.1038/srep44990.; Frenette P. S., Weiss L. Sulfated glycans induce rapid hematopoietic progenitor cell mobilization: Evidence for selectin-dependent and independent mechanisms. Blood. 2000; 96 (7): 2460–8. doi:10.1182/blood.v96.7.2460.h8002460_2460_2468.; Kubonishi S., Kikuchi T., Yamaguchi S., Tamamura H., Fujii N., Watanabe T., Arenzana-Seisdedos F., Ikeda K., Matsui T., Tanimoto M., Katayama Y. Rapid hematopoietic progenitor mobilization by sulfated colominic acid. Biochem Biophys Res Commun. 2007; 355 (4): 970–5. doi:10.1016/j.bbrc.2007.02.069.; Anisimova N. Y., Ustyuzhanina N. E., Bilan M. I., Donenko F. V., Ushakova N. A., Usov A. I., Kiselevskiy M. V., Nifantiev N. E. Infl uence of modified fucoidan and related sulfated oligosaccharides on hematopoiesis in cyclophosphamide-induced mice. Mar Drugs. 2018; 16 (9): 333. doi:10.3390/md16090333.; Yan H., Baldridge M. T., King K. Y. Hematopoiesis and the bacterial microbiome. Blood. 2018; 132 (6): 559–64. doi:10.1182/blood-2018-02-832519.; Iwamura C., Bouladoux N., Belkaid Y., Sher A., Jankovic D. Sensing of the microbiota by NOD1 in mesenchymal stromal cells regulates murine hematopoiesis. Blood. 2017; 129 (2): 171–6. doi:10.1182/blood-2016-06-723742.; Staffas A., Burgos da Silva M., Slingerland A. E., Lazrak A., Bare C. J., Holman C. D., Docampo M. D., Shono Y., Durham B., Pickard A. J., Cross J. R., Stein-Thoeringer C., Velardi E., Tsai J. J., Jahn L., Jay H., Lieberman S., Smith O. M., Pamer E. G., Peled J. U., Cohen D. E., Jenq R. R., van den Brink M. R. M. Nutritional support from the intestinal microbiota improves hematopoietic reconstitution after bone marrow transplantation in mice. Cell Host Microbe. 2018; 23 (4): 447–57.e4. doi:10.1016/j.chom.2018.03.002.; Peled J. U., Devlin S. M., Staffas A., Lumish M., Khanin R., Littmann E. R., Ling L., Kosuri S., Maloy M., Slingerland J. B., Ahr K. F., Porosnicu Rodriguez K. A., Shono Y., Slingerland A. E., Docampo M. D., Sung A. D., Weber D, Alousi A. M., Gyurkocza B., Ponce D. M., Barker J. N., Perales M. A., Giralt S. A., Taur Y., Pamer E. G., Jenq R. R., van den Brink M. R. M. Intestinal microbiota and relapse after hematopoietic-cell transplantation. J Clin Oncol. 2017; 35 (15): 1650–9. doi:10.1200/JCO.2016.70.3348.; Shang Q., Shan X., Cai C. Dietary fucoidan modulates the gut microbiota in mice by increasing the abundance of Lactobacillus and Ruminococcaceae. Food Funct. 2016; 7 (7): 3224–32. doi:10.1039/c6fo00309e.; Cumashi A., Ushakova N. A., Preobrazhenskaya M. E., D'Incecco A., Piccoli A., Totani L., Tinari N., Morozevich G. E., Berman A. E., Bilan M. I., Usov A. I., Ustyuzhanina N. E., Grachev A. A., Sanderson C. J., Kelly M., Rabinovich G. A., Iacobelli S., Nifantiev N. E. Consorzio Interuniversitario Nazionale per la Bio-Oncologia, Italy. Comparative study of the anti-inflammatory, anticoagulant, antiangiogenic, and antiadhesive activities of nine different fucoidans from brown seaweeds. Glycobiology. 2007; 17 (5): 541–52. doi:10.1093/glycob/cwm014.; Pomin V. H. Holothurian fucosylated chondroitin sulfates. Mar Drugs. 2014; 12 (1): 232–54. doi:10.3390/md12010232.; Ustyuzhanina N. E., Ushakova N. A., Zyuzina K. A., Bilan M. I., Elizarova A. L., Somonova O. V., Madzhuga A. V., Krylov V. B., Preobrazhenskaya M. E., Usov A. I., Kiselevskiy M. V., Nifantiev N. E. Influence of fucoidans on hemostatic system. Mar Drugs. 2013; 11 (7): 2444–58. doi:10.3390/md11072444.; https://journal.nodgo.org/jour/article/view/884
-
3Academic Journal
المؤلفون: K. A. Sergeenko, E. B. Machneva, T. Z. Aliev, I. O. Kostareva, T. T. Valiev, M. V. Kiselevskiy, R. Ya. Vlasenko, K. I. Kirgizov, S. R. Varfolomeeva, К. А. Сергеенко, Е. Б. Мачнева, Т. З. Алиев, И. О. Костарева, Т. Т. Валиев, М. В. Киселевский, Р. Я. Власенко, К. И. Киргизов, С. Р. Варфоломеева
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 10, № 1 (2023); 57-67 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 10, № 1 (2023); 57-67 ; 2413-5496 ; 2311-1267
مصطلحات موضوعية: Элизария, transplant-associated thrombotic microangiopathy, eculizumab, mesenchymal stem cells, Elizaria, трансплантат-ассоциированная тромботическая микроангиопатия, экулизумаб, мезенхимальные стволовые клетки
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/922/816; Ditschkowski M., Elmaagacli A., Trenschel R., Gromke T., Steckel N., Koldehoff M., Beelen D. Dynamic international prognostic scoring system scores, pre-transplant therapy and chronic graft-versus-host disease determine outcome after allogeneic hematopoietic stem cell transplantation for myelofibrosis. Haematologica. 2012;97:1574–81. doi:10.3324/haematol.2011.061168.; Saudemont A., Madrigal J.A. Allogeneic T cells: Maestro in the co-ordination of the immune response after hematopoietic stem cell transplantation. Haematologica. 2014;99:203–5. doi:10.3324/haematol.2013.101295.; Corre E., Carmagnat M., Busson M., Peffault de Latour R., Robin M., Ribaud P., Toubert A., Rabian C., Socié G. Long-term immune deficiency after allogeneic stem cell transplantation: B-cell deficiency is associated with late infections. Haematologica. 2010;95:1025–9. doi:10.3324/haematol.2009.018853.; Moake J.L., Byrnes J.J. Thrombotic microangiopathies associated with drugs and bone marrow transplantation. Hematol Oncol Clin North Am. 1996;10(2):485–97. doi:10.1016/s0889-8588(05)70348-8.; George J.N., Li X., McMinn J., Terrell D., Vesely S., Selby G. Thrombotic thrombocytopenic purpurahemolytic uremic syndrome following allogeneic HPC transplantation: a diagnostic dilemma. Transfusion. 2004;44(2):294–304. doi:10.1111/j.1537-2995.2004.00700.x.; Dervenoulas J., Tsirigotis P., Bollas G., Pappa V., Xiros N., Economopoulos T., Pappa M., Mellou S., Kostourou A., Papageorgiou E., Raptis S.A. Thrombotic thrombocytopenic purpura/ hemolytic uremic syndrome (TTP/HUS): treatment outcome, relapses, prognostic factors. A single-center experience of 48 cases. Ann Hematol. 2000;79(2):66–72. doi:10.1007/s002770050012.; Ruggenenti P., Noris M., Remuzzi G. Thrombotic microangiopathies. In: Wilcox C.S., editor. Therapy in nephrology & hypertension: a companion to Brenner & Rector’s the kidney. 3rd edition. Philadelphia: Saunders, 2008. Pp. 294–312.; Ariceta G., Besbas N., Johnson S., Karpman D., Landau D., Licht C., Loirat C., Pecoraro C., Taylor M., van de Kar N., Vandewalle J., Zimmerhackl L.B. Guideline for the investigation and initial therapy of diarrhea-negative hemolytic uremic syndrome. Pediatr Nephrol. (Berlin, Germany) 2009;24(4):687–96. doi:10.1007/s00467-008-0964-1.; Willems E., Baron F., Seidel L., Frère P., Fillet G., Beguin Y. Comparison of thrombotic microangiopathy after allogeneic hematopoietic cell transplantation with high-dose or nonmyeloablative conditioning. Bone Marrow Transplant. 2010;45:689–93. doi:10.1038/bmt.2009.230.; Rosenthal J., Powlowska A., Bolotin E., Cervantes C., Maroongroge S., Thomas S., Forman S.J. Transplant-associated thrombotic microangiopathy in pediatric patients treated with sirolimus and tacrolimus. Pediatr Blood Cancer. 2011;57:142–6. doi:10.1002/pbc.22861.; Rodriguez R., Nakamura R., Palmer J.M., Parker P., Shayani S., Nademanee A., Snyder D., Pullarkat V., Kogut N., Rosenthal J., Smith E., Karanes C., O’Donnell M., Krishnan A., Senitzer D., Forman S.J. A phase II pilot study of tacrolimus/sirolimus GVHD prophylaxis for sibling donor hematopoietic stem cell transplantation using 3 conditioning regimens. Blood. 2010;115:1098–105. doi:10.1182/blood-2009-03-207563.; Changsirikulchai S., Myerson D., Guthrie K.A., McDonald G.B., Alpers C.E., Hingorani S.R. Renal thrombotic microangiopathy after hematopoietic cell transplant: Role of GVHD in pathogenesis. Clin J Am Soc Nephrol. 2009;4:345–53. doi:10.2215/CJN.02070508.; Biedermann B.C. Vascular endothelium and graft-versus-host disease. Best Pract Res Clin Haematol. 2008;21:129–38. doi:10.1016/j.beha.2008.02.003.; George J.N., Selby G.B. Thrombotic microangiopathy after allogeneic bone marrow transplantation: a pathologic abnormality associated with diverse clinical syndromes. Bone Marrow Transplant. 2004;33:1073–4. doi:10.1038/sj.bmt.1704513.; Ho V., Cutler C., Carter S., Martin P., Adams R., Horowitz M., Ferrara J., Soiffer R., Giralt S. Blood and Marrow Transplant Clinical Trials Network Toxicity Committee consensus summary: thrombotic microangiopathy after hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2005;11:571–5. doi:10.1016/j.bbmt.2005.06.001.; Batts E.D., Lazarus H.M. Diagnosis and treatment of transplantationassociated thrombotic microangiopathy: Real progress or are we still waiting? Bone Marrow Transplant. 2007;40:709–19. doi:10.1038/sj.bmt.1705758.; Chang A., Hingorani S., Kowalewska J., Flowers M.E.D., Aneja T., Smith K.D., Meehan S.M., Nicosia R.F., Alpers C.E. Spectrum of renal pathology in hematopoietic cell transplantation: a series of 20 patients and review of the literature. Clin J Am Soc Nephrol. 2007;2:1014–23. doi:10.2215/CJN.01700407.; Uderzo C., Bonanomi S., Busca A., Renoldi M., Ferrari P., Iacobelli M., Morreale G., Lanino E., Annaloro C., Volpe A.D., Alessandrino P., Longoni D., Locatelli F., Sangalli H., Rovelli A. Risk factors and severe outcome in thrombotic microangiopathy after allogeneic hematopoietic stem cell transplantation. Transplantation. 2006;82:638–44. doi:10.1097/01.tp.0000230373.82376.46.; Lopes da Silva R., Ferreira I., Teixeira G., Cordeiro D., Mafra M., Costa I., Bravo Marques J.M., Abecasis M. BK virus encephalitis with thrombotic microangiopathy in an allogeneic hematopoietic stem cell transplant recipient. Transpl Infect Dis. 2011;13:161–7. doi:10.1111/j.1399-3062.2010.00581.x.; Nürnberger W., Michelmann I., Burdach S., Göbel U. Endothelial dysfunction after bone marrow transplantation: Increase of soluble thrombomodulin and PAI-1 in patients with multiple transplant related complications. Ann Hematol. 1998;76:61–5. doi:10.1007/s002770050364.; Takatsuka H., Takemoto Y., Yamada S., Wada H., Tamura S., Fujimori Y., Okamoto T., Suehiro A., Kanamaru A., Kakishita E. Complications after bone marrow transplantation are manifestations of systemic inflammatory response syndrome. Bone Marrow Transplant. 2000;26:419–26. doi:10.1038/sj.bmt.1702517.; Jodele S., Dandoy C.E., Myers K.C., El-Bietar J., Nelson A., Wallace G., Laskin B.L. New approaches in the diagnosis, pathophysiology, and treatment of pediatric hematopoietic stem cell transplantation-associated thrombotic microangiopathy. Transfus Apher Sci. 2016;54(2):181–90. doi:10.1016/j.transci.2016.04.007.; Jodele S., Laskin B.L., Dandoy C.E., Myers K.C., El-Bietar J., Davies S.M., Goebel J., Dixon B. A new paradigm: diagnosis and management of HSCT-associated thrombotic microangiopathy as multi-system endothelial injury. Blood Rev. 2014;29(3):191–204. doi:10.1016/j.blre.2014.11.001.; Young J.A., Pallas C.R., Knovich M.A. Transplant-associated thrombotic microangiopathy: theoretical considerations and a practical approach to an unrefined diagnosis. Bone Marrow Transplant. 2021;56(8):1805–17. doi:10.1038/s41409-021-01283-0.; Jodele S., Dandoy C.E., Lane A., Laskin B.L., Teusink-Cross A., Myers K.C. Complement blockade for TA-TMA: lessons learned from a large pediatric cohort treated with eculizumab. Blood. 2020;135:1049–57. doi:10.1182/blood.2019004218.; Jodele S., Fukuda T., Vinks A., Mizuno K., Laskin B.L., Goebel J. Eculizumab therapy in children with severe hematopoietic stem cell transplantation-associated thrombotic microangiopathy. Biol Blood Marrow Transplant. 2014;20:518–25. doi:10.1016/j.bbmt.2013.12.565.; Jodele S., Fukuda T., Mizuno K., Vinks A.A., Laskin B.L., Goebel J. Variable eculizumab clearance requires pharmacodynamic monitoring to optimize therapy for thrombotic microangiopathy after hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2016;22:307–15. doi:10.1016/j.bbmt.2015.10.002.; Khaled S.K., Kwong Y.L., Smith M., Metjian A., Whitaker S. Early Results of Phase II Study Using OMS721 in Patients with Hematopoietic Stem Cell Transplant-Associated Thrombotic Microangiopathy (HCT-TMA). Biol Blood Marrow Transplant. 2017;23:S282–3. doi:10.1016/j.bbmt.2016.12.192.; Carreras E., Dufour C., Mohty M., Kröger N. Тhe EBMT Handbook: Hematopoietic Stem Cell Transplantation and Cellular Therapies, 2019. 688 р.; Aversa F., Terenzi A., Tabilio A., Falzetti F., Carotti A., Ballanti S. Full haplotype-mismatched hematopoietic stem-cell transplantation: a phase II study in patients with acute leukemia at high risk of relapse. J Clin Oncol. 2005;23:3447–54. doi:10.1200/JCO.2005.09.117.; Luznik L., O’Donnell P.V., Symons H.J., Chen A.R., Leffell M.S., Zahurak M., Gooley T.A., Piantadosi S., Kaup M., Ambinder R.F., Huff C.A., Matsui W., Bolaños-Meade J., Borrello I., Powell J.D., Harrington E., Warnock S., Flowers M., Brodsky R.A., Sandmaier B.M., Storb R.F., Jones R.J., Fuchs E.J. HLA-haploidentical bone marrow transplantation for hematologic malignancies using nonmyeloablative conditioning and high-dose, posttransplantation cyclophosphamide. Biol Blood Marrow Transplant. 2008;14:641–50. doi:10.1016/j.bbmt.2008.03.005.; Wang Y., Liu D.H., Liu K.Y., Xu L.P., Zhang X.H., Han W. Long-term follow-up of haploidentical hematopoietic stem cell transplantation without in vitro T cell depletion for the treatment of leukemia: nine years of experience at a single center. Cancer. 2013;119:978–85. doi:10.1002/cncr.27761.; Olsson R.F., Logan B.R., Chaudhury S., Zhu X., Akpek G., Bolwell B.J. Primary graft failure after myeloablative allogeneic hematopoietic cell transplantation for hematologic malignancies. Leukemia. 2015;29:1754–62. doi:10.1038/leu.2015.75.; Uccelli A., Moretta L., Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol. 2008;8:726–36. doi:10.1038/nri2395.; Muguruma Y., Yahata T., Miyatake H., Tadayuki S., Tomoko U., Jobu I., Shunichi K., Mamoru I., Tomomitsu H., Kiyoshi A. Reconstitution of the functional human hematopoietic microenvironment derived from human mesenchymal stem cells in the murine bone marrow compartment. Blood. 2006;107:1878–87. doi:10.1182/blood-2005-06-2211.; Kiel M.J., Morrison S.J. Uncertainty in the niches that maintain haematopoietic stem cells. Nat Rev Immunol. 2008;8:290–301. doi:10.1038/nri2279.; Скоробогатова Е.В., Киргизов К.И., Пристанскова Е.А., Благонравова О.Л., Осипова Е.Ю., Пономарева Н.И., Пурбуева Б.Б., Буря А.Е., Константинова В.В., Филина О.А., Мачнева Е.Б., Ольхова Л.В., Мезенцева А.В., Антошин М.М., Сидорова Н.В. Внутрикостное введение мезенхимальных стволовых клеток в терапии рефрактерной реакции «трансплантат против хозяина» с гипофункцией трансплантата. Педиатрия. Журнал им. Г.Н. Сперанского. 2019;98(4):8–14.; Muroi K., Miyamura K., Ohashi K. Unrelated allogeneic bone marrow-derived mesenchymal stem cells for steroid-refractory acute graft-versus-host disease: a phase I/II study. Int J Hematol. 2013;98:206–13. doi:10.1007/s12185-013-1399-4.; Muroi K., Miyamura K., Okada M., Yamashita T., Murata M., Ishikawa T., Uike N., Hidaka M., Kobayashi R., Imamura M., Tanaka J., Ohashi K., Taniguchi S., Ikeda T., Eto T., Mori M., Yamaoka M., Ozawa K. Bone marrow-derived mesenchymal stem cells (JR-031) for steroid-refractory grade III or IV acute graft-versushost disease: a phase II/III study. Int J Hematol. 2016;103:243–50. doi:10.1007/s12185-015-1915-9.; Ankrum J.A., Ong J.F., Karp J.M. Mesenchymal stem cells: immune evasive, not immune privileged. Nat Biotechnol. 2014;32:252–60. doi:10.1038/nbt.2816.; Kulagin A., Ptushkin V., Lukina E., Davydkin I., Korobkin A., Shamrai V., Konstantinova T., Kaporskaya T., Mitina T., Ksenzova T., Zuev E., Markova O., Gapchenko E., Kudlay D. Randomized multicenter noninferiority phase III clinical trial of the first biosimilar of eculizumab. Ann Hematol. 2021;100(11):2689–98. doi:10.1007/s00277-021-04624-7.; Kulagin A., Ptushkin V., Lukina E., Gapchenko E., Markova O., Zuev E., Kudlay D. Phase III clinical trial of Elizaria® and Soliris® in adult patients with paroxysmal nocturnal hemoglobinuria: results of comparative analysis of effi cacy, safety, and pharmacological data. Blood. 2019;134(Suppl. 1):3748. doi:10.1182/blood-2019-125693.; Птушкин В.В., Кулагин А.Д., Лукина Е.А., Давыдкин И.Л., Константинова Т.С., Шамрай В.С., Минаева Н.В., Кудлай Д.А., Гапченко Е.В., Маркова О.А., Борозинец А.Ю. Результаты открытого многоцентрового клинического исследования Ib фазы по оценке безопасности, фармакокинетики и фармакодинамики первого биоаналога экулизумаба у нелеченых пациентов с пароксизмальной ночной гемоглобинурией в фазе индукции терапии. Терапевтический архив. 2020;92(7):77–84. doi:10.26442/00403660. 2020.07.000818.; Мачнева Е.Б., Болохонова М.А., Алиев Т.З., Шевцов Д.В., Сулейманова А.М., Сидорова Н.В., Османов Е.А., Киргизов К.И., Варфоломеева С.Р. Тромботическая микроангиопатия, ассоциированная с трансплантацией гемопоэтических стволовых клеток: общая характеристика и пример из клинической практики. Российский журнал детской гематологии и онкологии. 2020;7(3):86–93. doi:10.21682/2311-1267-2020-7-3-86-93.; Эмирова Х.М., Орлова О.М., Музуров А.Л., Генералова Г.А., Панкратенко Т.Е., Абасеева Т.Ю., Шаталов П.А., Козина А.А., Ильинский В.В., Шустер А.М., Кудлай Д.А. Опыт применения Элизарии® при атипичном гемолитико-уремическом синдроме. Педиатрия. Журнал им. Г.Н. Сперанского. 2019;98(5):225–9.; Rasmusson I., Ringdén O., Sundberg B., Le Blanc K. Mesenchymal stem cells inhibit the formation of cytotoxic T lymphocytes, but not activated cytotoxic T lymphocytes or natural killer cells. Transplantation. 2003;76:1208–13. doi:10.1097/01.TP.0000082540.43730.80.; Kiselevskiy M., Vlasenko R., Reshetnikova V., Chikileva I., Shubina I., Osmanov E., Valiev T., Sidorova N., Batmanova N., Stepanyan N., Kirgizov K., Varfolomeeva S. Potential use of mesenchymal multipotent cells for hemopoietic stem cell transplantation: pro and contra. J Pediatr Hematol Oncol. 2021;43(3):90–4. doi:10.1097/MPH.0000000000002065.; Киселевский М.В., Власенко Р.Я., Степанян Н.Г., Шубина И.Ж., Ситдикова С.М., Киргизов К.И., Варфоломеева С.Р. Секрет мезенхимных стволовых клеток костного мозга: иммуносупрессивный или провоспалительный? Клеточные технологии в биологии и медицине. 2021;3:171–5. doi:10.47056/1814-3490-2021-3-171-175.; https://journal.nodgo.org/jour/article/view/922
-
4Academic Journal
المؤلفون: K. A. Sergeenko, T. Z. Aliev, I. O. Kostareva, Yu. V. Lozovan, M. D. Malova, N. A. Batmanova, T. T. Valiev, E. B. Machneva, K. I. Kirgizov, К. А. Сергеенко, Т. З. Алиев, И. О. Костарева, Ю. В. Лозован, М. Д. Малова, Н. А. Батманова, Т. Т. Валиев, Е. Б. Мачнева, К. И. Киргизов
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 9, № 2 (2022); 85-92 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 9, № 2 (2022); 85-92 ; 2413-5496 ; 2311-1267
مصطلحات موضوعية: блинатумомаб, inotuzumab ozogamicin, venoocclusive disease, minimal residual disease, blinatumomab, инотузумаб озогамицин, веноокклюзионная болезнь, минимальная остаточная болезнь
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/833/738; Bunn H.F., Aster J.C. Acute Leukemias. In: Pathophysiology of Blood Disorders. The McGraw-Hill Companies, Inc., 2011. Pp. 244–259.; Bhojwani D., Pui C.H. Relapsed childhood acute lymphoblastic leukaemia. Lancet Oncol. 2013;14:e205–17. doi:10.1016/S1470-2045(12)70580-6.; Sun W., Malvar J., Sposto R., Verma A., Wilkes J.J., Dennis R. Outcome of children with multiply relapsed B-cell acute lymphoblastic leukemia: a therapeutic advances in childhood leukemia & lymphoma study. Leukemia. 2018;32(11):2316–25. doi:10.1038/s41375-018-0094-0.; Ko R.H., Ji L., Barnette P., Bostrom B., Hutchinson R., Raetz E. Outcome of patients treated for relapsed or refractory acute lymphoblastic leukemia: a Therapeutic Advances in Childhood Leukemia Consortium study. J Clin Oncol. 2010;28:648–54. doi:10.1200/JCO.2009.22.2950.; Dahl J., Marx K., Jabbour E. Inotuzumab ozogamicin in the treatment of acute lymphoblastic leukemia. Expert Rev Hematol. 2016;9:329–34. doi:10.1586/17474086.2016.1143771.; Tedder T.F., Poe J.C., Haas K.M. CD22: a multifunctional receptor that regulates B lymphocyte survival and signal transduction. Adv Immunol. 2005;88:1–50. doi:10.1016/S0065-2776(05)88001-0.; Shah N.N., Stevenson M.S., Yuan C.M., Richards K., Delbrook C., Kreitman R.J. Characterization of CD22 expression in acute lymphoblastic leukemia. Pediatr Blood Cancer. 2015;62:964–9. doi:10.1002/pbc.25410.; DeAngelo D.J., Stock W., Stein A.S., Shustov A., Liedtke M., Schiff er C.A. Inotuzumab ozogamicin in adults with relapsed or refractory CD22-positive acute lymphoblastic leukemia: a phase 1/2 study. Blood Adv. 2017;1:1167–80. doi:10.1182/bloodadvances.2016001925.; Kantarjian H., Thomas D., Jorgensen J., Jabbour E., Kebriaei P., Rytting M. Inotuzumab ozogamicin, an anti-CD22- calecheamicin conjugate, for refractory and relapsed acute lymphocytic leukaemia: a phase 2 study. Lancet Oncol. 2012;13:403–11. doi:10.1016/S1470-2045(11)70386-2.; Benjamin J.E., Stein A.S. The role of blinatumomab in patients with relapsed/refractory acute lymphoblastic leukemia. Ther Adv Hematol. 2016;7(3):142–56. doi:10.1177/2040620716640422.; Locatelly F., Whitlock J., Peters C., Chen-Santel C., Chia V., Dennis R.M., Heym K.M., Katz A.J., Kelsh M.A., Sposto R., Tu H., Tuglus C.A., Verma A., Vinti L., Wilkes J.J., Zubarovskaja N., Zugmaier G., von Stackelberg A., Sun W. Blinatumomab versus historical standard therapy in pediatric patients with relapsed/refractory Ph-negative B-cell precursor acute lymphoblastic leukemia. Leukemia. 2020;34(9):2473–8. doi:10.1038/s41375-020-0770-8.; Батманова Н.А., Валиев Т.Т., Киргизов К.И., Варфоломеева С.Р. Применение блинатумомаба в терапии острого лимфобластного лейкоза у детей: особенности организации терапии, обеспечения препаратом и токсичность лечения. Обзор литературы и собственный опыт. Российский журнал детской гематологии и онкологии. 2021;8(4):39–46. doi:10.21682/2311-1267-2021-8-4-39-46.; Hoff man L.M., Gore L. Blinatumomab, a bi-specific anti-CD19/CD3 BiTE® antibody for the treatment of acute lymphoblastic leukemia: perspectives and current pediatric applications. Front Oncol. 2014;4:63. doi:10.3389/fonc.2014.00063.; Nagorsen D., Kufer P., Baeuerle P.A., Bargou R. Blinatumomab: a historical perspective. Pharmacol Ther. 2012;136(3):334–42. doi:10.1016/j.pharmthera.2012.07.013.; von Stackelberg A., Locatelli F., Zugmaier G., Handgretinger R., Trippett T.M., Rizzari C., Bader P., OʼBrien M.M., Brethon B., Bhojwani D., Schlegel P.G., Borkhardt A., Rheingold S.R., Cooper T.M., Zwaan C.M., Barnette P., Messina C., Michel G., DuBois S.G., Hu K., Zhu M., Whitlock J.A., Gore L. Phase I/Phase II Study of Blinatumomab in Pediatric Patients With Relapsed/Refractory Acute Lymphoblastic Leukemia. J Clin Oncol. 2016;34(36):4381–9. doi:10.1200/JCO.2016.67.3301.; Gokbuget N., Zugmaier G., Klinger M., Kufer P., Stelljes M., Viardot A., Horst H.A., Neumann S., Brüggemann M., Ottmann O.G., Burmeister T., Wessiepe D., Topp M.S., Bargou R. Long-term relapsefree survival in a phase 2 study of blinatumomab for the treatment of patients with minimal residual disease in B-lineage acute lymphoblastic leukemia. Haematologica. 2017;102(4):e132–5. doi:10.3324/haematol.2016.153957.; Jasinski S., De Los Reyes F., Yametti G.C., Pierro J., Raetz E., Caroll W.L. Immunotherapy in pediatric B-acute lymphoblastic leukemia: Advances and ongoing challenges. Paediatr Drugs. 2020;22(5):485–99. doi:10.1007/s40272-020-00413-3.; Yurkiewicz I.R., Muffl y L., Liedtke M. Inotuzumab ozogamicin: a CD22 mAb-drug conjugate for adult relapsed or refractory B-cell precursor acute lymphoblastic leukemia. Drug Des Devel Ther. 2018;12:2293–300. doi:10.2147/DDDT.S150317.; DiJoseph J.F., Armellino D.C., Boghaert E.R., Khandke K., Dougher M.M., Sridharan L., Kunz A., Hamann P.R., Gorovits B., Udata C., Moran J.K., Popplewell A.G., Stephens S., Frost P., Damle N.K. Antibody-targeted chemotherapy with CMC-544: a CD22-targeted immunoconjugate of calicheamicin for the treatment of B-lymphoid malignancies. Blood. 2004;103(5):1807–14. doi:10.1182/blood-2003-07-2466.; Shor B., Gerber H.P., Sapra P. Preclinical and clinical development of inotuzumab-ozogamicin in hematological malignancies. Mol Immunol. 2015;67:107–16. doi:10.1016/j.molimm.2014.09.014.; Boue D.R., LeBien T.W. Expression and structure of CD22 in acute leukemia. Blood. 1988;71(5):1480–6. PMID: 3258772.; Thota S., Advani A. Inotuzumab ozogamicin in relapsed B-cell acute lymphoblastic leukemia. Eur J Haematol. 2017;98(5):425–34. doi:10.1111/ejh.12862.; Haso W., Lee D.W., Shah N.N., Stetler-Stevenson M., Yuan C.M., Pastan I.H., Dimitrov D.S., Morgan R.A., FitzGerald D.J., Barrett D.M., Wayne A.S., Mackall C.L., Orentas R.J. Anti-CD22-chimeric antigen receptors targeting B-cell precursor acute lymphoblastic leukemia. Blood. 2013;121(7):1165–74. doi:10.1182/blood-2012-06-438002.; De Vries J.F., Zwaan C.M., De Bie M., Voerman J.S.A., den Boer M.L., van Dongen J.J.M., van der Velden V.H.J. The novel calicheamicin-conjugated CD22 antibody inotuzumab ozogamicin (CMC-544) eff ectively kills primary pediatric acute lymphoblastic leukemia cells. Leukemia. 2012;26(2):255–64. doi:10.1038/leu.2011.206.; Giles F.J., Kantarjian H.M., Kornblau S.M., Thomas D.A., Garcia-Manero G., Waddelow T.A., David C.L., Phan A.T., Colburn D.E., Rashid A., Estey E.H. Mylotarg (gemtuzumab ozogamicin) therapy is associated with hepatic venoocclusive disease in patients who have not received stem cell transplantation. Cancer. 2001;92(2):406–13. doi:10.1002/10970142(20010715)92:23.0.co;2-u.; McKoy J.M., Angelotta C., Bennett C.L., Tallman M.S., Wadleigh M., Evens A.M., Kuzel T.M., Trifi lio S.M., Raisch D.W., Kell J., DeAngelo D.J., Giles F.J. Gemtuzumab ozogamicin-associated sinusoidal obstructive syndrome (SOS): an overview from the research on adverse drug events and reports (RADAR) project. Leuk Res. 2007;31(5):599–604. doi:10.1016/j.leukres.2006.07.005.; Godwin C.D., McDonald G.B., Walter R.B. Sinusoidal obstruction syndrome following CD33-targeted therapy in acute myeloid leukemia. Blood. 2017;129(16):2330–2. doi:10.1182/blood-2017-01-762419.; Kantarjian H.M., DeAngelo D.J., Stelljes M., Martinelli G., Liedtke M., Stock W., Gökbuget N., O’Brien S., Wang K., Wang T., Paccagnella M.L., Sleight B., Vandendries E., Advani A.S. Inotuzumab ozogamicin versus standard therapy for acute lymphoblastic leukemia. N Engl J Med. 2016;375:740–53. doi:10.1056/NEJMoa1509277.; Pfi zer Ltd. BESPONSA™ (inotuzumab ozogamicin) summary of product characteristics, 2020.; Lamb Y.N. Inotuzumab Ozogamicin: First Global Approval. Drugs. 2017;77(14):1603–10. doi:10.1007/s40265-017-0802-5.; Rytting M., Triche L., Thomas D. Initial experience with CMC544 (inotuzumab ozogamicin) in pediatric patients with relapsed B cell acute lymphoblastic leukemia. Pediatr Blood. 2014;544:369–72. doi:10.1002/pbc.24721.; Bhojwani D., Sposto R., Shah N.N., Rodriguez V., Yuan C., Stetler-Stevenson M., O’Brien M.M., McNeer J.L., Quereshi A., Cabannes A., Schlegel P., Rossig C., Dalla-Pozza L., August K., Alexander S., Bourquin J.-P., Zwaan M., Raetz E.A., Loh M.L., Rheingold S.R. Inotuzumab ozogamicin in pediatric patients with relapsed/refractory acute lymphoblastic leukemia. Leukemia. 2018;33(4):884–92. doi:10.1038/s41375-018-0265-z.; https://journal.nodgo.org/jour/article/view/833
-
5
المؤلفون: I. O. Kostareva, K. I. Kirgizov, E. B. Machneva, T. Z. Aliev, Yu. V. Lozovan, K. A. Sergeenko, N. A. Burlaka, T. I. Potemkina, K. V. Mitrakov, A. Yu. Yelfimova, A. S. Slinin, M. D. Malova, R. R. Fatkhullin, N. G. Stepanyan, N. A. Batmanova, T. T. Valiev, S. R. Varfolomeeva
المصدر: Pediatric Hematology/Oncology and Immunopathology. 22:16-23
مصطلحات موضوعية: Oncology, Immunology, Pediatrics, Perinatology and Child Health, Immunology and Allergy, Hematology
-
6Academic Journal
المؤلفون: N. V. Sidorova, E. B. Machneva, T. T. Valiev, I. O. Kostareva, T. Z. Aliev, A. Yu. Elfimova, Yu. V. Lozovan, T. Yu. Pavlova, Yu. S. Korkina, T. V. Gorbunova, Z. V. Grigorievskaya, I. N. Petukhova, N. Yu. Epifanova, K. I. Kirgizov, Н. В. Сидорова, Е. Б. Мачнева, Т. Т. Валиев, И. О. Костарева, Т. З. Алиев, А. Ю. Елфимова, Ю. В. Лозован, Т. Ю. Павлова, Ю. С. Коркина, Т. В. Горбунова, З. В. Григорьевская, И. Н. Петухова, Н. Ю. Епифанова, К. И. Киргизов
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 8, № 2 (2021); 61-72 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 8, № 2 (2021); 61-72 ; 2413-5496 ; 2311-1267
مصطلحات موضوعية: цефтазидим/авибактам, hematopoietic stem cell transplantation, Klebsiella pneumoniae, multidrug resistance, ceftazidime/avibactam, трансплантация гемопоэтических стволовых клеток, множественная лекарственная устойчивость
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/721/660; Скоробогатова Е.В., Балашов Д.Н., Трахтман П.Е., Масчан А.А., Румянцев А.Г., Киргизов К.И. Итоги двадцатилетнего опыта трансплантации гемопоэтических стволовых клеток у детей. Педиатрия. Журнал им. ГН. Сперанского 2011;90(4):12-6.; Афанасьев Б.В., Зубаровская Л.С., Моисеев И.С. Аллогенная трансплантация гемопоэтических стволовых клеток у детей: настоящее, проблемы и перспективы. Российский журнал детской гематологии и онкологии 2015;2(2):28-42. doi:10.17650/2311-1267-2015-2-228-42.; Multani A., Allard L.S., Wangjam T., Sica R.A., Epstein D.J., Rezvani A.R., Ho D.Y. Missed diagnosis and misdiagnosis of infectious diseases in hematopoietic cell transplant recipients: an autopsy study. Blood Adv 2019;3(22):3602-12. doi:10.1182/bloodadvances.2019000634.; Ge J., Yang T., Zhang L., Zhang X., Zhu X., Tang B., Wan X., Tong J., Song K., Yao W., Sun G., Sun Z., Liu H. The incidence, risk factors and outcomes of early bloodstream infection in patients with malignant hematologic disease after unrelated cord blood transplantation: a retrospective study. BMC Infect Dis 2018;18(1):654. doi:10.1186/s12879-018-3575-x.; Mikulska M., Del Bono V., Bruzzi P., Raiola A. M., Gualandi F., Van Lint M.T., Bacigalupo A., Viscoli C. Mortality after bloodstream infections in allogeneic haematopoietic stem cell transplant (HSCT) recipients. Infection 2012;40(3):271-8. doi:10.1007/s15010-011-0229-y.; Wang L., Wang Y., Fan X., Tang W., Hu J. Prevalence of Resistant Gram-Negative Bacilli in Bloodstream Infection in Febrile Neutropenia Patients Undergoing Hematopoietic Stem Cell Transplantation: A Single Center Retrospective Cohort Study. Medicine (Baltimore) 2015;94(45):e1931. doi:10.1097/MD.0000000000001931.; Riccardi N., Rotulo G.A., Castagnola E. Definition of Opportunistic Infections in Immunocompromised Children on the Basis of Etiologies and Clinical Features: A Summary for Practical Purposes. Curr Pediatr Rev 2019;15(4):197-206. doi:10.2174/1573396315666190617151745.; Heinz W.J., Buchheidt D., Christopeit M., von Lilienfeld-Toal M., Cornely O.A., Einsele H., Karthaus M., Link H., Mahlberg R., Neumann S., Ostermann H., Penack O., Ruhnke M., Sandherr M., Schiel X., Vehreschild J.J., Weissinger F., Maschmeyer G. Diagnosis and empirical treatment of fever of unknown origin (FUO) in adult neutropenic patients: guidelines of the Infectious Diseases Working Party (AGIHO) of the German Society of Hematology and Medical Oncology (DGHO). Ann Hematol 2017;96(11):1775-92. doi:10.1007/s00277-017-3098-3.; Mikulska M., Viscoli C., Orasch C., Livermore D.M., Averbuch D., Cordonnier C., Akova M.; Fourth European Conference on Infections in Leukemia Group (ECIL-4), a joint venture of EBMT, EORTC, ICHS, ELN and ESGICH/ESCMID. Aetiology and resistance in bacteraemias among adult and paediatric haematology and cancer patients. J Infect 2014;68(4):321-31. doi:10.1016/j.jinf.2013.12.006.; Girmenia C., Bertaina A., Piciocchi A., Perruccio K., Algarotti A., Busca A., Cattaneo C., Raiola A.M., Guidi S., Iori A.P., Candoni A., Irrera G., Milone G., Marcacci G., Scime R., Musso M., Cudillo L., Sica S., Castagna L., Corradini P., Marchesi F., Pastore D., Alessandrino E.P., Annaloro C., Ciceri F., Santarone S., Nassi L., Farina C., Viscoli C., Rossolini G.M., Bonifazi F., Rambaldi A.; Gruppo Italiano Trapianto di Midollo Osseo (GITMO) and Associazione Microbiologi Clinici Italiani (AMCLI). Incidence, Risk Factors and Outcome of Pre-engraftment Gram-Negative Bacteremia After Allogeneic and Autologous Hematopoietic Stem Cell Transplantation: An Italian Prospective Multicenter Survey. Clin Infect Dis 2017;65(11):1884-96. doi:10.1093/cid/cix690.; Martm-Pena A., Aguilar-Guisado M., Espigado I., Parody R., Miguel Cisneros J. Prospective study of infectious complications in allogeneic hematopoietic stem cell transplant recipients. Clin Transplant 2011;25(3):468-74. doi:10.1111/j.1399-0012.2010.01286.x.; Gomez S., Fynn A.B., Fernanda S., Cecilia V., Sung L. Early bacterial and fungal infection in children receiving allogeneic stem cell transplantation for acute lymphoblastic leukemia in Argentina. Pediatr Transplant 2018;22(1). doi:10.1111/petr.13070.; Freifeld A.G., Bow E.J., Sepkowitz K.A., Boeckh M.J., Ito J.I., Mullen C.A., Raad I.I., Rolston K.V., Young J.A., Wingard J.R.; Infectious Diseases Society of America. Clinical practice guideline for the use of antimicrobial agents in neutropenic patients with cancer: 2010 update by the infectious diseases society of america. Clin Infect Dis 2011;52(4):e56-93. doi:10.1093/cid/cir073.; Averbuch D., Tridello G., Hoek J., Mikulska M., Akan H., Yanez San Segundo L., Pabst T., Oz^elik T., Klyasova G., Donnini I., Wu D., Gulbas Z., Zuckerman T., Botelho de Sousa A., Beguin Y., Xhaard A., Bachy E., Ljungman P., de la Camara R., Rascon J., Ruiz Camps I., Vitek A., Patriarca F., Cudillo L., Vrhovac R., Shaw P.J., Wolfs T., O'Brien T., Avni B., Silling G., Al Sabty F., Graphakos S., Sankelo M., Sengeloev H., Pillai S., Matthes S., Melanthiou F., Iacobelli S., Styczynski J., Engelhard D., Cesaro S. Antimicrobial Resistance in Gram-Negative Rods Causing Bacteremia in Hematopoietic Stem Cell Transplant Recipients: Intercontinental Prospective Study of the Infectious Diseases Working Party of the European Bone Marrow Transplantation Group. Clin Infect Dis 2017;65(11):1819-28. doi:10.1093/cid/cix646.; Forcina A., Baldan R., Marasco V., Cichero P., Bondanza A., Noviello M., Piemontese S., Soliman C., Greco R., Lorentino F., Giglio F., Messina C., Carrabba M., Bernardi M., Peccatori J., Moro M., Biancardi A., Nizzero P., Scarpellini P., Cirillo D.M., Mancini N., Corti C., Clementi M., Ciceri F. Control of infectious mortality due to carbapenemase-producing Klebsiella pneumoniae in hematopoietic stem cell transplantation. Bone Marrow Transplant 2017;52(1):114-9. doi:10.1038/bmt.2016.234.; Sadowska-Klasa A., Piekarska A., Prejzner W., Bieniaszewska M., Hellmann A. Colonization with multidrug-resistant bacteria increases the risk of complications and a fatal outcome after allogeneic hematopoietic cell transplantation. Ann Hematol 2018;97(3):509-17. doi:10.1007/s00277-017-3205-5.; Scheich S., Lindner S., Koenig R., Reinheimer C., Wichelhaus T.A., Hogardt M., Besier S., Kempf V.A.J., Kessel J., Martin H., Wilke A.C., Serve H., Bug G., Steffen B. Clinical impact of colonization with multidrug-resistant organisms on outcome after allogeneic stem cell transplantation in patients with acute myeloid leukemia. Cancer 2018;124(2):286-96. doi:10.1002/cncr.31045.; Forcina A., Lorentino F., Marasco V., Oltolini C., Marcatti M., Greco R., Lupo-Stanghellini M.T., Carrabba M., Bernardi M., Peccatori J., Corti C., Ciceri F. Clinical Impact of Pretransplant Multidrug-Resistant Gram-Negative Colonization in Autologous and Allogeneic Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2018;24(7):1476-82. doi:10.1016/j.bbmt.2018.02.021.; Averbuch D., Cordonnier C., Livermore D.M., Mikulska M., Orasch C., Viscoli C., Gyssens I.C., Kern W.V., Klyasova G., Marchetti O., Engelhard D., Akova M.; ECIL4, a joint venture of EBMT, EORTC, ICHS, ESGICH/ESCMID and ELN. Targeted therapy against multiresistant bacteria in leukemic and hematopoietic stem cell transplant recipients: guidelines of the 4th European Conference on Infections in Leukemia (ECIL-4, 2011). Haematologica 2013;98(12):1836-47. doi:10.3324/haematol.2013.091330.; Mazuski J.E., Gasink L.B., Armstrong J., Broadhurst H., Stone G.G., Rank D., Llorens L., Newell P., Pachl J. Efficacy and Safety of Ceftazidime-Avibactam Plus Metronidazole Versus Meropenem in the Treatment of Complicated Intra-abdominal Infection: Results From a Randomized, Controlled, Double-Blind, Phase 3 Program. Clin Infect Dis 2016;62(11):1380-9. doi:10.1093/cid/ciw133.; Козлов Р.С., Стецюк О.У., Андреева И.В. Цефтазидим-авибактам: новые «правила игры» против полирезистентных грамотрицательных бактерий. Клиническая микробиология и антимикробная химиотерапия 2018;20(1):24-34. doi:10.36488/cmac.2018.1.24-34.; Caston J.J., Lacort-Peralta I., Martm-Davila P., Loeches B., Tabares S., Temkin L., Torre-Cisneros J., Pano-Pardo J.R. Clinical efficacy of ceftazidime/avibactam versus other active agents for the treatment of bacteremia due to carbapenemase-producing Enterobacteriaceae in hematologic patients. Int J Infect Dis 2017;59:118-23. doi:10.1016/j.ijid.2017.03.021.; Shaw E., Rombauts A., Tubau F., Padulles A., Camara J., Lozano T., Cobo-Sacristan S., Sabe N., Grau I., Rigo-Bonnin R., Dominguez M.A., Carratala J. Clinical outcomes after combination treatment with ceftazidime/avibactam and aztreonam for NDM-1/OXA-48/CTX-M-15-producing Klebsiella pneumoniae infection. J Antimicrob Chemother 2018;73(4):1104-6. doi:10.1093/jac/dkx496.; Singer M., Deutschman C.S., Seymour C.W., Shankar-Hari M., Annane D., Bauer M., Bellomo R., Bernard G.R., Chiche J.D., Coopersmith C.M., Hotchkiss R.S., Levy M.M., Marshall J.C., Martin G.S., Opal S.M., Rubenfeld G.D., van der Poll T., Vincent J.L., Angus D.C. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 2016;315(8):801-10. doi:10.1001/jama.2016.0287.; Satlin M.J., Cohen N., Ma K.C., Gedrimaite Z., Soave R., Askin G., Chen L., Kreiswirth B.N., Walsh T.J., Seo S.K. Bacteremia due to carbapenem-resistant Enterobacteriaceae in neutropenic patients with hematologic malignancies. J Infect 2016;73(4):336-45. doi:10.1016/j.jinf.2016.07.002.; Mokart D., Slehofer G., Lambert J., Sannini A., Chow-Chine L., Brun J.P., Berger P., Duran S., Faucher M., Blache J.L., Saillard C., Vey N., Leone M. De-escalation of antimicrobial treatment in neutropenic patients with severe sepsis: results from an observational study. Intensive Care Med 2014;40(1):41-9. doi:10.1007/s00134-013-3148-9.; Orasch C., Averbuch D., Mikulska M., Cordonnier C., Livermore D.M., Gyssens I.C., Klyasova G., Engelhard D., Kern W., Viscoli C., Akova M., Marchetti O.; 4th European Conference on Infections in Leukemia (ECIL-4); joint venture of Infectious Diseases Working Party of the European Group for Blood and Marrow Transplantation (IDWP-EBMT); Infectious Diseases Group of the European Organization for Research and Treatment of Cancer (IDG-EORTC); International Immunocompromised Host Society (ICHS); European Leukemia Net (ELN) and European Study Group on Infections in Immunocompromised Hosts of the European Society for Clinical Microbiology and Infectious Diseases (ESGICH-ESCMID). Discontinuation of empirical antibiotic therapy in neutropenic leukaemia patients with fever of unknown origin is ethical. Clin Microbiol Infect 2015;21(3):e25-7. doi:10.1016/j.cmi.2014.10.014.; D'Amico F., Soverini M., Zama D., Consolandi C., Severgnini M., Prete A., Pession A., Barone M., Turroni S., Biagi E., Brigidi P., Masetti R., Rampelli S., Candela M. Gut resistome plasticity in pediatric patients undergoing hematopoietic stem cell transplantation. Sci Rep 2019;9(1):5649. doi:10.1038/s41598-019-42222-w.; Karaiskos I., Giamarellou H. Multidrug-resistant and extensively drugresistant Gram-negative pathogens: current and emerging therapeutic approaches. Expert Opin Pharmacother 2014;15(10):1351-70. doi:10.1517/14656566.2014.914172.; Averbuch D., Orasch C., Cordonnier C., Livermore D.M., Mikulska M., Viscoli C., Gyssens I.C., Kern W.V., Klyasova G., Marchetti O., Engelhard D., Akova M.; ECIL4, a joint venture of EBMT, EORTC, ICHS, ESGICH/ESCMID and ELN. European guidelines for empirical antibacterial therapy for febrile neutropenic patients in the era of growing resistance: summary of the 2011 4th European Conference on Infections in Leukemia. Haematologica 2013;98(12):1826-35. doi:10.3324/haematol.2013.091025. Erratum in: Haematologica 2014;99(2):400. PMID: 24323983.; https://journal.nodgo.org/jour/article/view/721
-
7Academic Journal
المؤلفون: T. Z. Aliev, T. S. Belysheva, K. A. Sergeenko, E. B. Machneva, N. V. Sidorova, T. T. Valiev, I. O. Kostareva, K. I. Kirgizov, Т. З. Алиев, Т. С. Белышева, К. А. Сергеенко, Е. Б. Мачнева, Н. В. Сидорова, Т. Т. Валиев, И. О. Костарева, К. И. Киргизов
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 8, № 3 (2021); 97-101 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 8, № 3 (2021); 97-101 ; 2413-5496 ; 2311-1267
مصطلحات موضوعية: треосульфан, hematopoietic stem cell transplantation, damage, toxicity, Jelonet, treosulfan, трансплантация гемопоэтических стволовых клеток, поражение, токсичность
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/748/681; Rosman I.S., Lloyd B.M., Hayashi R.J., Bayliss S.J. Cutaneous effects of thiotepa in pediatric patients receiving high-dose chemotherapy with autologous stem cell transplantation. J Am Acad Dermatol 2008;58(4):575–8. doi:10.1016/j.jaad.2007.12.037.; The EBMT Handbook. Hematopoietic Stem Cell Transplantation and Cellular Therapies. Eds.: Carreras E., Dufour C., Mohty M., Kröger N. Springer, 2019. 688 р. doi:10.1007/978-3-030-02278-5; Mays S.R., Kunishige J.H., Truong E., Kontoyiannis D.P., Hymes S.R. Approach to the morbilliform eruption in the hematopoietic transplant patient. Semin Cutan Med Surg 2007;26(3):155–62. doi:10.1016/j.sder.2007.09.004.; Strocchio L., Zecca M., Comoli P., Mina T., Giorgiani G., Giraldi E., Vinti L., Merli P., Regazzi M., Locatelli F. Treosulfan-based conditioning regimen for allogeneic haematopoietic stem cell transplantation in children with sickle cell disease. Br J Haematol 2015;169(5):726–36. doi:10.1111/bjh.13352.; Bolognia J.L., Cooper D.L., Glusac E.J. Toxic erythema of chemotherapy: a useful clinical term. J Am Acad Dermatol 2008;59(3):524–9. doi:10.1016/j.jaad.2008.05.018.; Алиев Т.З., Мачнева Е.Б. Сидорова Н.В., Белышева Т.С., Валиев Т.Т., Киргизов К.И. Поражение кожных покровов при трансплантации гемопоэтических стволовых клеток. Обзор литературы. Вопросы гематологии/онкологии и иммунопатологии в педиатрии 2020;19(2):184–92. doi:10.24287/1726-1708-2020-19-2-184-192. [Aliev T.Z., Machneva E.B., Sidorova N.V., Belysheva T.S., Valiev T.T., Kirgizov K.I. Skin damage after the hematopoietic stem cell transplantation. Literature review. Voprosy gematologii/onkologii i immunopatologii v pediatrii = Pediatric Hematology/Oncology and Immunopathology 2020;19(2):184–92. (In Russ.)].; Руководство по эксплуатации от производителя Smith & Nephew Medical Limited, 101 Hessle Road, Hull HU3 2BN, England, 2015.; Foyatier J.L., Hezez G., Masson C.L., Latarjet J. Use of a neutral paraffin dressing (Jelonet) at the Burn Care Center at the Saint Luc Hospital in Lyon. Ann Chir Plast Esthet 1990;35(4):327–30. PMID: 1702958.; Rothenberger J., Constantinescu M.A., Held M., Aebersold D.M., Stolz A., Tschumi C., Olariu R. Use of a Polylactide-based Copolymer as a Temporary Skin Substitute for a Patient With Moist Desquamation Due to Radiation. Wounds 2016;28(7):E26–30. PMID: 27428721.; Schwarze H., Küntscher M., Uhlig C., Hierlemann H., Prantl L., Noack N., Hartmann B. Suprathel, a new skin substitute, in the management of donor sites of split-thickness skin grafts: results of a clinical study Burns 2007;33(7):850–4. doi:10.1016/j.burns.2006.10.393.; https://journal.nodgo.org/jour/article/view/748
-
8Academic Journal
المؤلفون: A. V. Mezentseva, L. V. Olkhova, E. B. Machneva, V. V. Konstantinova, A. E. Burya, Yu. A. Nikolaeva, O. A. Filina, B. B. Purbueva, O. S. Fink, M. M. Antoshin, T. Z. Aliev, A. V. Martynenkova, E. A. Pristanskova, N. V. Sidorova, K. I. Kirgizov, E. V. Skorobogatova, А. В. Мезенцева, Л. В. Ольхова, Е. Б. Мачнева, В. В. Константинова, А. Е. Буря, Ю. А. Николаева, О. А. Филина, Б. Б. Пурбуева, О. С. Финк, М. М. Антошин, Т. З. Алиев, А. В. Мартыненкова, Е. А. Пристанскова, Н. В. Сидорова, К. И. Киргизов, Е. В. Скоробогатова
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 8, № 1 (2021); 23-34 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 8, № 1 (2021); 23-34 ; 2413-5496 ; 2311-1267
مصطلحات موضوعية: дети, hydroxymethylquinoxaline dioxide (dioxidine), Pseudomonas aeruginosa, Klebsiella pneumoniae, Enterobacter cloacae, Stenotrophomonas maltophilia, children, гидроксиметилхиноксалиндиоксид (диоксидин)
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/687/630; Никифорова Г.Н., Свистушкин В.М., Дедова М.Г. Возможности местной терапии инфекционно-воспалительных заболеваний ЛОР-органов. Русский медицинский журнал 2015;6:346–9.; Падейская Е.Н. Антибактериальный препарат диоксидин: особенности биологического действия и значение в терапии различных форм гнойной инфекции. Инфекции и антимикробная терапия 2001;3(5):150–5.; Падейская Е.Н. Антимикробные препараты в ряду производных сульфаниламида, диаминопиримидина, 5-нитроимидазола ди-N-оксихиноксалина. Русский медицинский журнал 1997;21:1–6.; Попов Д.А., Анучкина Н.М., Терентьев А.А., Костюк Г.В., Блатун Л.А., Русанова Е.В., Александрова И.А., Пхакадзе Т.Я., Богомолова Н.С., Терехова Л.П. Диоксидин: антимикробная активность и перспективы клинического применения на современном этапе. Антибиотики и химиотерапия 2013;58:3–4. PMID: 24640151.; Дурнев А.Д., Дубовская О.Ю., Нигарова Э.А., Середенин С.Б. Роль свободных радикалов кислорода в механизме мутагенного действия диоксидина. Химико-фармацевтический журнал 1989;23(11):1289–91.; Suter W., Russelet A., Knüsel F. Mode of Action of Quindoxin and Substituted quinoxaline-di-N-oxides on Escherichia Coli. Antimicrob Agents Chemother 1978;13(5):770–83. doi:10.1128/aac.13.5.770.; Отчет о результатах оценки активности in vitro антимикробного препарата диоксидин на базах НЦССХ им. А.Н. Бакулева, при участии НИИ нейрохирургии им. акад. Н.Н. Бурденко, НИИ хирургии им. А.В. Вишневского, ЦИТО им. Н.Н. Приорова, МОНИКИ им. М.Ф. Владимирского.; Падейская Е.Н., Шипилова Л.Д., Буданова Л.И. Фармакокинетика диоксидина, проникновение препарата в органы и ткани при однократном и повторном введении. Химико-фармацевтический журнал 1983;6:667–71.; Мезенцева А.В., Ольхова Л.В., Мачнева Е.Б., Константинова В.В., Буря А.Е., Николаева Ю.А., Филина О.А., Пристанскова Е.А., Пурбуева Б.Б., Финк О.С., Антошин М.М., Скоробогатова Е.В. Терапия фульминантных инфекционных осложнений, вызванных грамотрицательными возбудителями, у пациентов с индуцированной аплазией кроветворения. Педиатрия 2020;99(4):252–7. doi:10.24110/0031-403X-2020-99-4-252-257.; https://journal.nodgo.org/jour/article/view/687
-
9Academic Journal
المؤلفون: E. B. Machneva, V. Yu. Panarina, T. Z. Aliev, D. V. Shevtsov, A. M. Suleymanova, V. V. Konstantinova, A. E. Burya, N. G. Stepanyan, Yu. V. Skvortsova, N. V. Sidorova, E. A. Osmanov, K. I. Kirgizov, S. R. Varfolomeeva, Е. Б. Мачнева, В. Ю. Панарина, Т. З. Алиев, Д. В. Шевцов, А. М. Сулейманова, В. В. Константинова, А. Е. Буря, Н. Г. Степанян, Ю. В. Скворцова, Н. В. Сидорова, Е. А. Османов, К. И. Киргизов, С. Р. Варфоломеева
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 7, № 2 (2020); 94-111 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 7, № 2 (2020); 94-111 ; 2413-5496 ; 2311-1267 ; 10.21682/2311-1267-2020-7-2
مصطلحات موضوعية: хроническая реакция «трансплантат против хозяина», immune reconstitution, immune system, immunosuppressive therapy, chronic “graft versus host” disease, иммунная реконституция, иммунная система, иммуносупрессивная терапия
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/608/555; Румянцев А.Г., Масчан А.А. Трансплантация гемопоэтических стволовых клеток у детей. М.: МИА, 2003. 910 с.; Cooke K.R., Luznik L., Sarantopoulos S., Hakim F.T., Jagasia M., Fowler D.H., van den Brink M.R.M., Hansen J.A., Parkman R., Miklos D.B., Martin P.J., Paczesny S., Vogelsang G., Pavletic S., Ritz J., Schultz K.R., Blazar B.R. The Biology of Chronic Graftversus- Host Disease: A Task Force Report from the National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease. Biol Blood Marrow Transplant 2017;23(2):211–34. doi:10.1016/j.bbmt.2016.09.023.; Cutler C., Logan B., Nakamura R., Johnston L., Choi S., Porter D., Hogan W.J., Pasquini M., MacMillan M.L., Hsu J.W., Waller E.K., Grupp S., McCarthy P., Wu J., Hu Z.H., Carter S.L., Horowitz M.M., Antin J.H. Tacrolimus/sirolimus vs tacrolimus/methotrexate as GVHD prophylaxis after matched, related donor allogeneic HCT. Blood 2014;124(8):1372–7. doi:10.1182/blood-2014-04-567164.; Kennedy G.A., Varelias A., Vuckovic S., Le Texier L., Gartlan K.H., Zhang P., Thomas G., Anderson L., Boyle G., Cloonan N., Leach J., Sturgeon E., Avery J., Olver S.D., Lor M., Misra A.K., Hutchins C., Morton A.J., Durrant S.T., Subramoniapillai E., Butler J.P., Curley C.I., MacDonald K.P.A., Tey S.K., Hill G.R. Addition of interleukin-6 inhibition with tocilizumab to standard graft-versushost disease prophylaxis after allogeneic stem-cell transplantation: a phase 1/2 trial. Lancet Oncol 2015;15(13):1451–9. doi:10.1016/S1470-2045(14)71017-4.; Скворцова Ю.В., Новичкова Г.А., Масчан А.А. Новое в патогенезе, диагностике и лечении хронической реакции «трансплантат против хозяина» после аллогенной ТГСК. Вопросы гематологии/ онкологии и иммунопатологии в педиатрии 2018;17(2):121–35. doi:10.24287/1726-1708-2018-17-2-121-135.; Jagasia M.H., Greinix H.T., Arora M., Williams K.M., Wolff D., Cowen E.W., Palmer J., Weisdorf D., Treister N.S., Cheng G.S., Kerr H., Stratton P., Duarte R.F., McDonald G.B., Inamoto Y., Vigorito A., Arai S., Datiles M.B., Jacobsohn D., Heller T., Kitko C.L., Mitchell S.A., Martin P.J., Shulman H., Wu R.S., Cutler C.S., Vogelsang G.B., Lee S.J., Pavletic S.Z., Flowers M.E. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: I. The 2014 Diagnosis and Staging Working Group report. Biol Blood Marrow Transplant 2015;21(3):389–401. doi:10.1016/j.bbmt. 2014.12.001.; Румянцев А.Г., Масчан А.А., Балашов Д.Н., Скворцова Ю.В. Федеральные клинические рекомендации по лечению хронической реакции «Трансплантат против хозяина» после трансплантации гемопоэтических стволовых клеток у детей. М., 2015. [Электронный ресурс]: http://www.nodgo.org/sites/default/fi les/29.(Г)хртгскР.pdf.; Holtick U., Albrecht M., Chemnitz J.M., Theurich S., Skoetz N., Scheid C., von Bergwelt-Baildon M. Bone marrow versus peripheral blood allogeneic haematopoietic stem cell transplantation for haematological malignancies in adults. Cochrane Database Syst Rev 2014;(4):CD010189. doi:10.1002/14651858.CD010189.pub2.; MacDonald K.P., Hill G.R., Blazar B.R. Chronic graft-versus-host disease: biological insights from preclinical and clinical studies. Blood 2017;129(1):13–21. doi:10.1182/blood-2016-06-686618.; Скворцова Ю.В., Балашов Д.Н., Делягин В.М. Болезнь трансплантат против хозяина после аллогенной трансплантации гемопоэтических стволовых клеток у детей. Медицинский Совет 2016;1(1):136–9. doi:10.21518/2079-701X-2016-1-136-139.; Arora M., Nagaraj S., Witte J., DeFor T.E., MacMillan M., Burns L.J., Weisdorf D.J. New classifi cation of chronic GVHD: added clarity from the consensus diagnoses. Bone Marrow Transplant 2009;43(2):149–53. doi:10.1038/bmt.2008.305.; Penack O., Marchetti M., Ruutu T., Aljurf M., Bacigalupo A., Bonifazi F., Ciceri F., Cornelissen J., Malladi R., Duarte R.F., Giebel S., Greinix H., Holler E., Lawitschka A., Mielke S., Mohty M., Arat M., Nagler A., Passweg J., Schoemans H., Socié G., Solano C., Vrhovac R., Zeiser R., Kröger N., Basak G.W. Prophylaxis and management of graft versus host disease after stem-cell transplantation for haematological malignancies: updated consensus recommendations of the European Society for Blood and Marrow Transplantation. Lancet Haematol 2020;7(2):e157–67. doi:10.1016/S2352-3026(19)30256-X.; McSweeney P.A., Niederwieser D., Shizuru J.A., Sandmaier B.M., Molina A.J., Maloney D.G., Chauncey T.R., Gooley T.A., Hegenbart U., Nash R.A., Radich J., Wagner J.L., Minor S., Appelbaum F.R., Bensinger W.I., Bryant E., Flowers M.E., Georges G.E., Grumet F.C., Kiem H.P., Torok-Storb B., Yu C., Blume K.G., Storb R.F. Hematopoietic cell transplantation in older patients with hematologic malignancies: replacing high-dose cytotoxic therapy with graft-versus-tumor eff ects. Blood 2001;97(11):3390–400. doi:10.1182/blood.v97.11.3390.; Ram R., Storer B., Mielcarek M., Sandmaier B.M., Maloney D.G., Martin P.J., Flowers M.E., Chua B.K., Rotta M., Storb R. Association between calcineurin inhibitor blood concentrations and outcomes after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant 2012;18(3):414–22. doi:10.1016/j.bbmt.2011.08.016.; Storb R., Deeg H.J., Whitehead J., Appelbaum F., Beatty P., Bensinger W., Buckner C.D., Clift R., Doney K., Farewell V., Hansen J., Hill R., Lum L., Martin P., McGuffi n R., Sanders J., Stewart P., Sullivan K., Witherspoon R., Yee G., Thomas D. Methotrexate and cyclosporine compared with cyclosporine alone for prophylaxis of acute graft versus host disease after marrow transplantation for leukemia. N Engl J Med 1986;314(12):729–35. doi:10.1056/NEJM198603203141201.; Gooptu M., Koreth J. Better acute graft-versus-host disease outcomes for allogeneic transplant recipients in the modern era: a tacrolimus eff ect? Haematologica 2017;102:806–8. doi:10.3324/haematol.2017.165266.; Hiraoka A., Ohashi Y., Okamoto S., Moriyama Y., Nagao T., Kodera Y., Kanamaru A., Dohy H., Masaoka T.; Japanese FK506 BMT (Bone Marrow Transplantation) Study Group. Phase III study comparing tacrolimus (FK506) with cyclosporine for graft-versushost disease prophylaxis after allogeneic bone marrow transplantation. Bone Marrow Transplant 2001;28:181–5. doi:10.1038/sj.bmt.1703097.; Inamoto Y., Flowers M.E., Appelbaum F.R., Carpenter P.A., Deeg H.J., Furlong T., Kiem H.P., Mielcarek M., Nash R.A., Storb R.F., Witherspoon R.P., Storer B.E., Martin P.J. A retrospective comparison of tacrolimus versus cyclosporine with methotrexate for immunosuppression after allogeneic hematopoietic cell transplantation with mobilized blood cells. Biol Blood Marrow Transplant 2011;17(7):1088–92. doi:10.1016/j.bbmt.2011.01.017.; Khoury H.J., Wang T., Hemmer M.T., Couriel D., Alousi A., Cutler C., Aljurf M., Antin J.H., Ayas M., Battiwalla M., Cahn J.Y., Cairo M., Chen Y.B., Gale R.P., Hashmi S., Hayashi R.J., Jagasia M., Juckett M., Kamble R.T., Kharfan-Dabaja M., Litzow M., Majhail N., Miller A., Nishihori T., Qayed M., Schoemans H., Schouten H.C., Socie G., Storek J., Verdonck L., Vij R., Wood W.A., Yu L., Martino R., Carabasi M., Dandoy C., Gergis U., Hematti P., Solh M., Jamani K., Lehmann L., Savani B., Schultz K.R., Wirk B.M., Spellman S., Arora M., Pidala J. Improved survival after acute graft-versus-host disease diagnosis in the modern era. Haematologica 2017;102(5):958–66. doi:10.3324/haematol.2016.156356.; Nash R.A., Antin J.H., Karanes C., Fay J.W., Avalos B.R., Yeager A.M., Przepiorka D., Davies S., Petersen F.B., Bartels P., Buell D., Fitzsimmons W., Anasetti C., Storb R., Ratanatharathorn V. Phase 3 study comparing methotrexate and tacrolimus with methotrexate and cyclosporine for prophylaxis of acute graft-versushost disease after marrow transplantation from unrelated donors. Blood 2000;96(6):2062–8. PMID: 10979948.; Ratanatharathorn V., Nash R.A., Przepiorka D., Devine S.M., Klein J.L., Weisdorf D., Fay J.W., Nademanee A., Antin J.H., Christiansen N.P., van der Jagt R., Herzig R.H., Litzow M.R., Wolff S.N., Longo W.L., Petersen F.B., Karanes C., Avalos B., Storb R., Buell D.N., Maher R.M., Fitzsimmons W.E., Wingard J.R. Phase III study comparing methotrexate and tacrolimus (prograf, FK506) with methotrexate and cyclosporine for graft-versus-host disease prophylaxis after HLA-identical sibling bone marrow transplantation. Blood 1998;92:2303–14. PMID: 9746768.; Kanda Y., Kobayashi T., Mori T., Tanaka M., Nakaseko C., Yokota A., Watanabe R., Kako S., Kakihana K., Kato J., Tanihara A., Doki N., Ashizawa M., Kimura S.I., Kikuchi M., Kanamori H., Okamoto S.; Kanto Study Group for Cell Therapy. A randomized controlled trial of cyclosporine and tacrolimus with strict control of blood concentrations after unrelated bone marrow transplantation. Bone Marrow Transplant 2016;51:103–9. doi:10.1038/bmt.2015.222.; Kharfan-Dabaja M., Mhaskar R., Reljic T., Pidala J., Perkins J.B., Djulbegovic B., Kumar A. Mycophenolate mofetil versus methotrexate for prevention of graft-versus-host disease in people receiving allogeneic hematopoietic stem cell transplantation. Cochrane Database Syst Rev 2014;7:CD010280. doi:10.1002/14651858.CD010280.pub2.; Hamilton B.K., Rybicki L., Dean R., Majhail N.S., Haddad H., Abounader D., Hanna R., Sobecks R., Duong H., Hill B.T., Copelan E., Bolwell B., Kalaycio M. Cyclosporine in combination with mycophenolate mofetil versus methotrexate for graft versus host disease prevention in myeloablative HLA-identical sibling donor allogeneic hematopoietic cell transplantation. Am J Hematol 2015;90:144–8. doi:10.1002/ajh.23882.; Ram R., Yeshurun M., Vidal L., Shpilberg O., Gafter-Gvili A. Mycophenolate mofetil vs. methotrexate for the prevention of graftversus- host-disease-systematic review and meta-analysis. Leuk Res 2014;38:352–60. doi:10.1016/j.leukres.2013.12.012.; Chhabra S., Liu Y., Hemmer M.T., Costa L., Pidala J.A., Couriel D.R., Alousi A.M., Majhail N.S., Stuart R.K., Kim D., Ringden O., Urbano-Ispizua A., Saad A., Savani B.N., Cooper B., Marks D.I., Socie G., Schouten H.C., Schoemans H., Abdel-Azim H., Yared J., Cahn J.Y., Wagner J., Antin J.H., Verdonck L.F., Lehmann L., Aljurf M.D., MacMillan M.L., Litzow M.R., Solh M.M., Qayed M., Hematti P., Kamble R.T., Vij R., Hayashi R.J., Gale R.P., Martino R., Seo S., Hashmi S.K., Nishihori T., Teshima T., Gergis U., Inamoto Y., Spellman S.R., Arora M., Hamilton B.K. Comparative analysis of calcineurin inhibitor-based methotrexate and mycophenolate mofetil-containing regimens for prevention of graftversus- host disease after reduced-intensity conditioning allogeneic transplantation. Biol Blood Marrow Transplant 2019;25:73–85. doi:10.1016/j.bbmt.2018.08.018.; Finke J., Bethge W.A., Schmoor C., Ottinger H.D., Stelljes M., Zander A.R., Volin L., Ruutu T., Heim D.A., Schwerdtfeger R., Kolbe K., Mayer J., Maertens J.A., Linkesch W., Holler E., Koza V., Bornhäuser M., Einsele H., Kolb H.J., Bertz H., Egger M., Grishina O., Socié G.; ATG-Fresenius Trial Group. Standard graftversus- host disease prophylaxis with or without anti-T-cell globulin in haematopoietic cell transplantation from matched unrelated donors: a randomised, open-label, multicentre phase 3 trial. Lancet Oncol 2009;10:855–64. doi:10.1016/S1470-2045(09)70225-6.; Finke J., Schmoor C., Bethge W.A., Ottinger H., Stelljes M., Volin L., Heim D., Bertz H., Grishina O., Socie G. Long-term outcomes after standard graft-versus-host disease prophylaxis with or without antihuman- T-lymphocyte immunoglobulin in haemopoietic cell transplantation from matched unrelated donors: fi nal results of a randomised controlled trial. Lancet Haematol 2017;4:e293–301. doi:10.1016/S2352-3026(17)30081-9.; Walker I., Panzarella T., Couban S., Couture F., Devins G., Elemary M., Gallagher G., Kerr H., Kuruvilla J., Lee S.J., Moore J., Nevill T., Popradi G., Roy J., Schultz K.R., Szwajcer D., Toze C., Foley R.; Canadian Blood and Marrow Transplant Group. Pretreatment with antithymocyte globulin versus no anti-thymocyte globulin in patients with haematological malignancies undergoing haemopoietic cell transplantation from unrelated donors: a randomised, controlled, open-label, phase 3, multicentre trial. Lancet Oncol 2016;17:164–73. doi:10.1016/S1470-2045(15)00462-3.; Kröger N., Solano C., Wolschke C., Bandini G, Patriarca F., Pini M., Nagler A., Selleri C., Risitano A., Messina G., Bethge W., Pérez de Oteiza J., Duarte R., Carella A.M., Cimminiello M., Guidi S., Finke J., Mordini N., Ferra C., Sierra J., Russo D., Petrini M., Milone G., Benedetti F., Heinzelmann M., Pastore D., Jurado M., Terruzzi E., Narni F., Völp A., Ayuk F., Ruutu T., Bonifazi F. Antilymphocyte globulin for prevention of chronic graft-versus-host disease. N Engl J Med 2016;374:43–53. doi:10.1056/NEJMoa1506002.; Bonifazi F., Solano C., Wolschke C., Sessa M., Patriarca F., Zallio F., Nagler A., Selleri C., Risitano A.M., Messina G., Bethge W., Herrera P., Sureda A., Carella A.M., Cimminiello M., Guidi S., Finke J., Sorasio R., Ferra C., Sierra J., Russo D., Benedetti E., Milone G., Benedetti F., Heinzelmann M., Pastore D., Jurado M., Terruzzi E., Narni F., Völp A., Ayuk F., Ruutu T., Kröger N. Acute GVHD prophylaxis plus ATLG after myeloablative allogeneic haemopoietic peripheral blood stem-cell transplantation from HLA-identical siblings in patients with acute myeloid leukaemia in remission: fi nal results of quality of life and long-term outcome analysis of a phase 3 randomised study. Lancet Haematol 2019;6:e89–99. doi:10.1016/S2352-3026(18)30214-X.; Rubio M.T., D’Aveni-Piney M., Labopin M., Hamladji R.M., Sanz M.A., Blaise D., Ozdogu H., Daguindeau E., Richard C., Santarone S., Irrera G., Yakoub-Agha I., Yeshurun M., Diez-Martin J.L., Mohty M., Savani B.N., Nagler A. Impact of in vivo T cell depletion in HLA-identical allogeneic stem cell transplantation for acute myeloid leukemia in fi rst complete remission conditioned with a fl udarabine iv-busulfan myeloablative regimen: a report from the EBMT Acute Leukemia Working Party. J Hematol Oncol 2017;10(1):31. doi:10.1186/s13045-016-0389-4.; Neumann T., Schneidewind L., Weigel M., Plis A., Vaizian R., Schmidt C.A., Krüger W. Ruxolitinib for Therapy of Graft-versus- Host Disease. Biomed. Res Int 2019;2019:8163780. doi:10.1155/2019/8163780.; Koc S., Leisenring W., Flowers M.E., Anasetti C., Deeg H.J., Nash R.A., Sanders J.E., Witherspoon R.P., Storb R., Appelbaum F.R., Martin P.J. Therapy for chronic graft-versus-host disease: a randomized trial comparing cyclosporine plus prednisone versus prednisone alone. Blood 2002;100:48–51. doi:10.1182/blood.v100.1.48.; Sullivan K.M., Witherspoon R.P., Storb R., Weiden P., Flournoy N., Dahlberg S., Deeg H.J., Sanders J.E., Doney K.C., Appelbaum F.R., McGuffi n R., McDonald G.B., Meyers J., Schubert M.M., Gauvreau J., Shulman H.M., Sale G.E., Anasetti C., Loughran T.P., Strom S., Nims J., Thomas E.D. Prednisone and azathioprine compared with prednisone and placebo for treatment of chronic graft-v-host disease: prognostic infl uence of prolonged thrombocytopenia after allogeneic marrow transplantation. Blood 1988;72:546–54. PMID: 3042041.; Arora M., Wagner J.E., Davies S.M., Blazar B.R., Defor T., Enright H., Miller W.J., Weisdorf D.F. Randomized clinical trial of thalidomide, cyclosporine, and prednisone versus cyclosporine and prednisone as initial therapy for chronic graft-versus-host disease. Biol Blood Marrow Transplant 2001;7:265–73. doi:10.1053/bbmt.2001.v7.pm11400948.; Yadav H., Peters S.G., Keogh K.A., Hogan W.J., Erwin P.J., West C.P., Kennedy C.C. Azithromycin for the Treatment of Obliterative Bronchiolitis after Hematopoietic Stem Cell Transplantation: A Systematic Review and Meta-Analysis. Biol Blood Marrow Transplant 2016;22(12):2264–9. doi:10.1016/j.bbmt.2016.08.027.; Williams K.M., Cheng G.S., Pusic I., Jagasia M., Burns L., Ho V.T., Pidala J., Palmer J., Johnston L., Mayer S., Chien J.W., Jacobsohn D.A., Pavletic S.Z., Martin P.J., Storer B.E., Inamoto Y., Chai X., Flowers M.E.D., Lee S.J. Fluticasone, Azithromycin, and Montelukast Treatment for New-Onset Bronchiolitis Obliterans Syndrome after Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant 2016;22(4):710–6. doi:10.1016/j.bbmt.2015.10.009.; Hakim A., Cooke K.R., Pavletic S.Z., Khalid M., Williams K.M., Hashmi S.K. Diagnosis and treatment of bronchiolitis obliterans syndrome accessible universally. Bone Marrow Transplant 2019;54:383–92. doi:10.1038/s41409-018-0266-6.; Bergeron A., Chevret S., Granata A., Chevallier P., Vincent L., Huynh A., Tabrizi R., Labussiere-Wallet H., Bernard M., Chantepie S., Bay J.O., Thiebaut-Bertrand A., Thepot S., Contentin N., Fornecker L.M., Maillard N., Risso K., Berceanu A., Blaise D., Peff ault de La Tour R., Chien J.W., Coiteux V., Socié G.; ALLOZITHRO Study Investigators. Eff ect of azithromycin on airfl ow decline-free survival after allogeneic hematopoietic stem cell transplant: the ALLOZITHRO randomized clinical trial. JAMA 2017;318:557–66. doi:10.1001/jama.2017.9938.; Zeiser R. Biology-driven developments in the therapy of acute graftversus- host disease. Hematology Am Soc Hematol Educ Program 2018;2018(1):236–41. DOI:10.1182/asheducation-2018.1.236.; Zeiser R., Blazar B.R. Preclinical models of acute and chronic graftversus- host disease: how predictive are they for a successful clinical translation? Blood 2016;127(25):3117–26. doi:10.1182/blood-2016-02-699082.; Escamilla Gomez V., Garcia-Gutiеrrez V., Lоpez Corral L., Garcia Cadenas I., Pеrez Martinez A., Mаrquez Malaver F.J., Caballero- Velаzquez T., Gonzаlez Sierra P.A., Viguria Alegria M.C., Parra Salinas I.M., Calderon Cabrera C., Gonzаlez Vicent M., Rodriguez Torres N., Parody Porras R., Ferra Coll C., Orti G., Valcаrcel Ferreiras D., De la Cаmara LLanzа R., Molеs P., Velаzquez-Kennedy K., João Mende M., Caballero Barrigоn D., Pеrez E., Martino Bofarull R., Saavedra Gerosa S., Sierra J., Poch M., Zudaire Ripa M.T., Diaz Pеrez M.A., Molina Angulo B., Sаnchez Ortega I., Sanz Caballer J., Montoro Gоmez J., Espigado Tocino I., Pеrez-Simоn J.A. Ruxolitinib in refractory acute and chronic graft-versus-host disease: a multicenter survey study. Bone Marrow Transplant 2020;55(3):641–8. doi:10.1038/s41409-019-0731-x.; Khoury H.J., Langston A.A., Kota V.K., Wilkinson J.A., Pusic I., Jillella A., Bauer S., Kim A.S., Roberts D., Al-Kadhimi Z., Bodo I., Winton E., Arellano M., DiPersio J.F. Ruxolitinib: a steroid sparing agent in chronic graft-versus-host disease. Bone Marrow Transplant 2018;53(7):826–31. doi:10.1038/s41409-017-0081-5.; Zeiser R., Burchert A., Lengerke C., Verbeek M., Maas-Bauer K., Metzelder S.K., Spoerl S., Ditschkowski M., Ecsedi M., Sockel K., Ayuk F., Ajib S., de Fontbrune F.S., Na I.K., Penter L., Holtick U., Wolf D., Schuler E., Meyer E., Apostolova P., Bertz H., Marks R., Lübbert M., Wäsch R., Scheid C., Stölzel F., Ordemann R., Bug G., Kobbe G., Negrin R., Brune M., Spyridonidis A., Schmitt-Gräff A., van der Velden W., Huls G., Mielke S., Grigoleit G.U., Kuball J., Flynn R., Ihorst G., Du J., Blazar B.R., Arnold R., Kröger N., Passweg J., Halter J., Sociе G., Beelen D., Peschel C., Neubauer A., Finke J., Duyster J., von Bubnoff N. Ruxolitinib in corticosteroidrefractory graft-versus-host disease after allogeneic stem cell transplantation: a multicenter survey. Leukemia 2015;29(10):2062–8. doi:10.1038/leu.2015.212.; https://journal.nodgo.org/jour/article/view/608
-
10Academic Journal
المؤلفون: T. Z. Aliev, K. I. Kirgizov, T. S. Belysheva, Т. З. Алиев, К. И. Киргизов, Т. С. Белышева
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 7, № 2 (2020); 112-114 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 7, № 2 (2020); 112-114 ; 2413-5496 ; 2311-1267 ; 10.21682/2311-1267-2020-7-2
مصطلحات موضوعية: кожные покровы, complications, care, children, skin, осложнения, уход, дети
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/609/556; Zimmer® Pulsavac® Plus. Семейство устройств для санации ране- вой поверхности. Информационный буклет. [Электронный ресурс]: http://www.zimmerrussia.ru/content/dam/zimmer-web/documents/ru-RU/pdf/medical-professionals/surgical Zimmer_Pulsavac_Plus_Family_BRO_RUS_corr.pdf; Jagasia M.H., Greinix H.T., Arora M., Williams K.M., Wolff D., Cowen E.W., Palmer J., Weisdorf D., Treister N.S., Cheng G.S., Kerr H., Stratton P., Duarte R.F., McDonald G.B., Inamoto Y., Vigorito A., Arai S., Datiles M.B., Jacobsohn D., Heller T., Kitko C.L., Mitchell S.A., Martin P.J., Shulman H., Wu R.S., Cutler C.S., Vogelsang G.B., Lee S.J., Pavletic S.Z., Flowers M.E. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: I. The 2014 Diagnosis and Staging Working Group report. Biol Blood Marrow Transplant 2015;21(3):389–401. doi:10.1016 / j.bbmt.2014.12.001.; Скворцова Ю.В., Новичкова Г.А., Масчан А.А. Новое в патогенезе, диагностике и лечении хронической реакции «трансплантат против хозяина» после аллогенной ТГСК. Вопросы гематологии/ онкологии и иммунопатологии в педиатрии 2018;17(2):121–35. doi:10.24287/1726-1708-2018-17-2-121-135.; https://journal.nodgo.org/jour/article/view/609
-
11Academic Journal
المؤلفون: E. B. Machneva, M. A. Bolokhonova, T. Z. Aliev, D. V. Shevtsov, A. M. Suleymanova, N. V. Sidorova, E. A. Osmanov, K. I. Kirgizov, Е. Б. Мачнева, М. А. Болохонова, Т. З. Алиев, Д. В. Шевцов, А. М. Сулейманова, Н. В. Сидорова, Е. А. Османов, К. И. Киргизов
المصدر: Russian Journal of Pediatric Hematology and Oncology; Том 7, № 3 (2020); 86-93 ; Российский журнал детской гематологии и онкологии (РЖДГиО); Том 7, № 3 (2020); 86-93 ; 2413-5496 ; 2311-1267 ; 10.21682/2311-1267-2020-7-3
مصطلحات موضوعية: реакция, thrombotic microangiopathy, graft versus host disease, calcineurin inhibitors, complement, rituximab, eculizumab, defibrotide, тромботическая микроангиопатия
وصف الملف: application/pdf
Relation: https://journal.nodgo.org/jour/article/view/630/576; Jodele S., Laskin B.L., Dandoy C.E., Myers K.C., El-Bietar J., Davies S.M., Goebel J., Dixon B.P. A new paradigm: Diagnosis and management of HSCT-associated thrombotic microangiopathy as multi-system endothelial injury. Blood Rev 2015;29(3):191‒204. doi:10.1016/j.blre.2014.11.001.; Carreras E., Dufour C., Mohty M., Kröger N. (eds.). The EBMT Handbook. Hematopoietic Stem Cell Transplantation and Cellular Therapies. This Springer, 2019. 688 р.; Scully M., Cataland S., Coppo P., de la Rubia J., Friedman K.D., Hovinga J.K., Lämmle B., Matsumoto M., Pavenski K., Sadler Е., Sarode R., Wu H.; International Working Group for Thrombotic Thrombocytopenic Purpura. Consensus on the standardization of terminology in thrombotic thrombocytopenic purpura and related thrombotic microangiopathies. J Thromb Haemost 2017;15:312–22. doi:10.1111/jth.13571.; Jodele S., Zhang K., Zou F., Laskin B., Dandoy C.E., Myers K.C., Lane А., Meller J., Medvedovic M., Chen J., Davies S.M. The genetic fingerprint of susceptibility for transplant-associated thrombotic microangiopathy. Blood 2016;127:989–96. doi:10.1182/blood-2015-08-663435.; Khosla J., Yeh A.C., Spitzer T.R., Dey B.R. Hematopoietic stem cell transplant-associated thrombotic microangiopathy: current paradigm and novel therapies. Bone Marrow Transplant 2018;53:129–37. doi:10.1038/bmt.2017.207.; Jodele S., Davies S.M., Lane A., Khoury J., Dandoy C., Goebel J., Myers K., Grimley M., Bleesing J., El-Bietar J., Wallace G., Chima R.S., Paff Z., Laskin B.L. Diagnostic and risk criteria for HSCT-associated thrombotic microangiopathy: a study in children and young adults. Blood 2014;124:645–53. doi:10.1182/blood-2014-03-564997.; Ho V.T., Cutler C., Carter S., Martin P., Adams R., Horowitz M., Ferrara J., Soiffer R., Giralt S. Blood and marrow transplant clinical trials network toxicity committee consensus summary: thrombotic microangiopathy after hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2005;11:571–5. doi:10.1016/j.bbmt.2005.06.001.; Ruutu T., Barosi G., Benjamin R.J., Clark R.E., George J.N., Gratwohl A., Holler E., Iacobelli M., Kentouche K., Lämmle B., Moake J.L., Richardson P., Socié G., Zeigler Z., Niederwieser D., Barbui T.; European Group for Blood and Marrow Transplantation; European LeukemiaNet. Diagnostic criteria for hematopoietic stem cell transplant-associated microangiopathy: results of a consensus process by an International Working Group. Haematologica 2007;92:95–100. doi:10.3324/haematol.10699.; Cho B.S., Yahng S.A., Lee S.E., Eom K.S., Kim Y.J., Kim H.J., Lee S., Min C.K., Cho G.S., Kim D.W., Lee J.W., Min W.S., Park C.W. Validation of recently proposed consensus criteria for thrombotic microangiopathy after allogeneic hematopoietic stem-cell transplantation. Transplantation 2010;90:918–26. doi:10.1097/tp.0b013e3181f24e8d.; Ruutu T., Hermans J., Niederwieser D., Gratwohl A., Kiehl M., Volin L., Bertz H., Ljungman P., Spence D., Verdonck L.F., Prentice H.G., Bosi A., du Toit C.E., Brinch L., Apperley J.F.; EBMT Chronic Leukaemia Working Party. Thrombotic thrombocytopenic purpura after allogeneic stem cell transplantation: a survey of the European Group for Blood and Marrow Transplantation (EBMT). Br J Haematol 2002;118:1112–9. doi:10.1046/j.1365-2141.2002.03721.x.; George J.N., Selby G.B. Thrombotic microangiopathy after allogeneic bone marrow transplantation: a pathologic abnormality associated with diverse clinical syndromes. Bone Marrow Transplant 2004;33:1073–4. doi:10.1038/sj.bmt.1704513.; Uderzo C.C., Jodele S., Missiry M.E., Ciceri F., Busca A., Bacigalupo A., Corbacioglu S. Transplant-associated thrombotic microangiopathy (TA-TMA) and consensus based diagnostic and therapeutic recommendations: which TA-TMA patients to treat and when? J Bone Marrow Res 2014;2:152–9. doi:10.4172/2329-8820.1000152.; Yeates L., Slatter M.A., Bonanomi S., Lim F.L.W.I., Ong S.Y., Dalissier A., Barberi W., Shulz A., Duval M., Heilmann C., Willekens A., Hwang W.H.Y., Uderzo C., Bader P., Gennery A.R. Use of defibrotide to treat transplant-associated thrombotic microangiopathy: a retrospective study of the paediatric diseases and inborn errors working parties of the European Society of Blood and Marrow Transplantation. Bone Marrow Transplant 2017;52:762–4. doi:10.1038/bmt.2016.351.; Jodele S., Fukuda T., Mizuno K., Vinks A.A., Laskin B.L., Goebel J., Dixon B.P., Chima R.S., Hirsch R, Teusink A., Lazear D., Lane A., Myers K.C., Dandoy C.E., Davies S.M. Variable Eculizumab Clearance Requires Pharmacodynamic Monitoring to Optimize Therapy for Thrombotic Microangiopathy after Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2016;22(2):307‒15. doi:10.1016/j.bbmt.2015.10.002.; Uderzo C., Bonanomi S., Busca A., Renoldi M., Ferrari P., Iacobelli M., Morreale G., Lanino E., Annaloro C., Della Volpe A., Alessandrino P., Longoni D., Locatelli F., Sangalli H., Rovelli A. Risk factors and severe outcome in thrombotic microangiopathy after allogeneic hematopoietic stem cell transplantation. Transplantation 2006;82:638‒44. doi:10.1097/01.tp.0000230373.82376.46.; Narimatsu H., Kami M., Hara S., Matsumura T., Miyakoshi S., Kusumi E., Kakugawa Y., Kishi Y., Murashige N., Yuji K., Masuoka K., Yoneyama A., Wake A., Morinaga S., Kanda Y. Intestinal thrombotic microangiopathy following reduced-intensity umbilical cord blood transplantation. Bone Marrow Transplant 2005;36:517‒23. doi:10.1038/sj.bmt.1705099.; Corti P., Uderzo C., Tagliabue A., Della Volpe A., Annaloro C., Tagliaferri E., Balduzzi A. Defibrotide as a promising treatment for thrombotic thrombocytopenic purpura in patients undergoing bone marrow transplantation. Bone Marrow Transplant 2002;29:542‒3. doi:10.1038/sj.bmt.1703414.; Davì G., Catalano I., Belvedere M., Amato S., Mogavero A., Giammarresi C., Alaimo P., Notarbartolo A. Effects of defibrotide on fibrinolytic activity in diabetic patients with stable angina pectoris. Thromb Res 1992;65:211‒20. doi:10.1016/0049-3848(92)90241-2.; Cella G., Sbarai A., Mazzaro G., Motta G., Carraro P., Andreozzi G.M., Hoppensteadt D.A., Fareed J. Tissue factor pathway inhibitor release induced by defibrotide and heparins. Clin Appl Thromb Hemost 2001;7:225‒8. doi:10.1177/107602960100700308.; Falanga A., Vignoli A., Marchetti M., Barbui T. Defibrotide reduces procoagulant activity and increases fibrinolytic properties of endothelial cells. Leukemia 2003;17:1636‒42. doi:10.1038/sj.leu.2403004.; Palmer K.J., Goa K.L. Defibrotide. A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic use in vascular disorders. Drugs 1993;45:259‒94. doi:10.2165/00003495-199345020-00007.; Laskin B.L., Goebel J., Davies S.M., Jodele S. Small vessels, big trouble in the kidneys and beyond: hematopoietic stem cell transplantation associated thrombotic microangiopathy. Blood 2011;118:1452‒62. doi:10.1182/blood-2011-02-321315.; Richardson P.G., Corbacioglu S., Ho V.T., Kernan N.A., Lehmann L., Maguire C., Maglio M., Hoyle M., Sardella M., Sergio G., Holler E., Carreras E., Niederwieser D., Soiffer R. Drug safety evaluation of defibrotide. Expert Opin Drug Saf 2013;12:123‒36. doi:10.1517/14740338.2012.749855.; Marr H., McDonald E.J., Merriman E., Smith M., Mangos H., Stoddart C., Ganly P. Successful treatment of transplant-associated microangiopathy with rituximab. N Z Med J 2009;122:72‒4. doi:10.1182/blood.V112.11.4337.4337.; Carella A.M., DʼArena G., Greco M.M., Nobile M., Cascavilla N. Rituximab for allo-SCT-associated thrombotic thrombocytopenic purpura. Bone Marrow Transplant 2008;41:1063‒5. doi:10.1038/bmt.2008.25.; Au W.Y., Ma E.S., Lee T.L., Ha S.Y., Fung A.T., Lie A.K.W., Kwong Y.L. Successful treatment of thrombotic microangiopathy after hematopoietic stem cell transplantation with rituximab. Br J Haematol 2007;137:475‒8. doi:10.1111/j.1365-2141.2007.06588.x.; Эмирова Х.М., Орлова О.М., Музуров А.Л., Генералова Г.А., Панкратенко Т.Е., Абасеева Т.Ю., Шаталов П.А., Козина А.А., Ильинский В.В., Шустер А.М., Кудлай Д.А. Опыт применения Элизарии® при атипичном гемолитико-уремическом синдроме. Педиатрия. Журнал им. Г.Н. Сперанского 2019;98(5):225‒9.; Птушкин В.В., Кулагин А.Д., Лукина Е.А., Давыдкин И.Л., Константинова Т.С., Шамрай В.С., Минаева Н.В., Кудлай Д.А., Гапченко Е.В., Маркова О.А., Борозинец А.Ю. Результаты открытого многоцентрового клинического исследования Ib фазы по оценке безопасности, фармакокинетики и фармакодинамики первого биоаналога экулизумаба у нелеченых пациентов с пароксизмальной ночной гемоглобинурией в фазе индукции терапии. Терапевтический архив 2020;92(7):77–84. doi:10.26442/00403660.2020.07.000818.; Kulagin A., Ptushkin V., Lukina E., Gapchenko E., Markova O., Zuev E., Kudlay D. Phase III clinical trial of Elizaria® and Soliris® in adult patients with paroxysmal nocturnal hemoglobinuria: results of comparative analysis of efficacy, safety, and pharmacological data. Blood 2019;134(Supp1):3748. doi: https://doi.org/10.1182/blood-2019-125693.; https://journal.nodgo.org/jour/article/view/630