يعرض 1 - 19 نتائج من 19 نتيجة بحث عن '"А. Л. Маслянский"', وقت الاستعلام: 0.57s تنقيح النتائج
  1. 1
    Academic Journal

    المصدر: Rheumatology Science and Practice; Vol 62, No 4 (2024); 431–434 ; Научно-практическая ревматология; Vol 62, No 4 (2024); 431–434 ; 1995-4492 ; 1995-4484

    وصف الملف: application/pdf

    Relation: https://rsp.mediar-press.net/rsp/article/view/3612/2394; Buttgereit F, Matteson EL, Dejaco C. Polymyalgia rheumatica and giant cell arteritis. JAMA. 2020;324(10):993-994. doi:10.1001/jama.2020.10155; Сатыбалдыев АМ, Демидова НВ, Савушкина НМ, Гордеев АВ. Ревматическая полимиалгия. Научно-практическая ревматология. 2018;56(2):215-227. doi:10.14412/1995-4484-2018215-227; Tomelleri A, van der Geest KSM, Khurshid MA, Sebastian A, Coath F, Robbins D, et al. Disease stratification in GCA and PMR: State of the art and future perspectives. Nat Rev Rheumatol. 2023;19(7):446-459. doi:10.1038/s41584-023-00976-8; Dejaco C, Kerschbaumer A, Aletaha D, Bond M, Hysa E, Camellino D, et al. Treat-to-target recommendations in giant cell arteritis and polymyalgia rheumatica. Ann Rheum Dis. 2024;83(1):48-57. doi:10.1136/ard-2022-223429; Hellmich B, Agueda A, Monti S, Buttgereit F, de Boysson H, Brouwer E, et al. 2018 update of the EULAR recommendations for the management of large vessel vasculitis. Ann Rheum Dis. 2020;79(1):19-30. doi:10.1136/annrheumdis-2019-215672; Toyoda T, Armitstead Z, Bhide S, Engamba S, Henderson E, Jones C, et al. Treatment of polymyalgia rheumatica: British Society for Rheumatology guideline scope. Rheumatol Adv Pract. 2024;8(1):rkae002. doi:10.1093/rap/rkae002; Matteson EL, Buttgereit F, Dejaco C, Dasgupta B. Glucocorticoids for management of polymyalgia rheumatica and giant cell arteritis. Rheum Dis Clin North Am. 2016;42(1):75-90viii. doi:10.1016/j.rdc.2015.08.009; Mainbourg S, Addario A, Samson M, Puéchal X, François M, Durupt S, et al. Prevalence of giant cell arteritis relapse in patients treated with glucocorticoids: A meta-analysis. Arthritis Care Res (Hoboken). 2020;72(6):838-849. doi:10.1002/acr.23901; Floris A, Piga M, Chessa E, Congia M, Erre GL, Angioni MM, et al. Long-term glucocorticoid treatment and high relapse rate remain unresolved issues in the real-life management of polymyalgia rheumatica: A systematic literature review and meta-analysis. Clin Rheumatol. 2022;41(1):19-31. doi:10.1007/s10067-021-05819-z; Nepal D, Putman M, Unizony S. Giant cell arteritis and polymyalgia rheumatica: Treatment approaches and new targets. Rheum Dis Clin North Am. 2023;49(3):505-521. doi:10.1016/j.rdc.2023.03.005; Насонов ЕЛ, Лила АМ. Ингибиция интерлейкина 6 при имму новоспалительных ревматических заболеваниях: достижения, перспективы и надежды. Научно-практическая ревматология. 2017;55(6):590-599. doi:10.14412/1995-4484-2017-590-599; Weyand CM, Goronzy JJ. Immunology of giant cell arteritis. Circ Res. 2023;132(2):238-250. doi:10.1161/CIRCRESAHA.122.322128; Kastrati K, Aletaha D, Burmester GR, Chwala E, Dejaco C, Dougados M, et al. A systematic literature review informing the consensus statement on efficacy and safety of pharmacological treatment with interleukin-6 pathway inhibition with biological DMARDs in immune-mediated inflammatory diseases. RMD Open. 2022;8(2):e002359. doi:10.1136/rmdopen-2022-002359; Насонов ЕЛ, Сатыбалдыев АМ, Оттева ЭН, Бекетова ТВ, Баранов АА. Фармакотерапия гигантоклеточного артериита и ревматической полимиалгии: перспективы применения моноклональных антител к интерлейкину 6. Научно-практическая ревматология. 2024;62(3):262-279. doi:10.47360/1995-4484-2024-262-279; Feist E, Nasonov E,. Interleukin 6 inhibition in rheumatoid arthritis: Highlight on olokizumab. Touch Reviews in RMD. 2023;2(1):17-27. doi:10.17925/RMD.2023.2.1.17; Dasgupta B, Cimmino MA, Maradit-Kremers H, Schmidt WA, Schirmer M, Salvarani C, et al. 2012 provisional classification criteria for polymyalgia rheumatica: A European League Against Rheumatism/American College of Rheumatology collaborative initiative. Ann Rheum Dis. 2012;71(4):484-492. doi:10.1136/ANNRHEUMDIS-2011-200329; Ponte C, Grayson PC, Robson JC, Suppiah R, Gribbons KB, Judge A, et al. 2022 American College of Rheumatology/EULAR classification criteria for giant cell arteritis. Ann Rheum Dis. 2022;81(12):1647-1653. doi:10.1136/ARD-2022-223480; https://rsp.mediar-press.net/rsp/article/view/3612

  2. 2
    Academic Journal

    المساهمون: Исследование выполнено при финансовой поддержке Министерства науки и высшего образования Российской Федерации (соглашение № 075-15-2022-301 от 20.04.2022)

    المصدر: Rheumatology Science and Practice; Vol 62, No 5 (2024); 501-512 ; Научно-практическая ревматология; Vol 62, No 5 (2024); 501-512 ; 1995-4492 ; 1995-4484

    وصف الملف: application/pdf

    Relation: https://rsp.mediar-press.net/rsp/article/view/3636/2404; Насонов ЕЛ (ред.). Ревматология. Российские клинические рекомендации. М.:ГЭОТАР-Медиа;2017.; Pasoto SG, Adriano de Oliveira Martins V, Bonfa E. Sjögren’s syndrome and systemic lupus erythematosus: Links and risks. Open Access Rheumatol. 2019;11:33-45. doi:10.2147/OARRR.S167783; Ambrus JL, Suresh L, Peck A. Multiple roles for B-lymphocytes in Sjogren’s syndrome. J Clin Med. 2016;5(10):87. doi:10.3390/jcm5100087; Groom J, Kalled SL, Cutler AH, Olson C, Woodcock SA, Schneider P, et al. Association of BAFF/BLyS overexpression and altered B cell differentiation with Sjögren’s syndrome. J Clin Invest. 2002;109(1):59-68. doi:10.1172/JCI14121; Nocturne G, Seror R, Fogel O, Belkhir R, Boudaoud S, Saraux A, et al. CXCL13 and CCL11 serum levels and lymphoma and disease activity in primary Sjögren’s syndrome. Arthritis Rheumatol. 2015;67(12):3226-3233. doi:10.1002/art.39315; Carvajal Alegria G, Gazeau P, Hillion S, Daïen CI, Cornec DYK. Could lymphocyte profiling be useful to diagnose systemic autoimmune diseases? Clin Rev Allergy Immunol. 2017;53(2):219-236. doi:10.1007/s12016-017-8608-5; Aringer M, Costenbader K, Daikh D, Brinks R, Mosca M, Ramsey-Goldman R, Smolen JS, et al. 2019 European League Against Rheumatism/American College of Rheumatology classification criteria for systemic lupus erythematosus. Arthritis Rheumatol. 2019;71(9):1400-1412. doi:10.1002/art.40930; Shiboski CH, Shiboski SC, Seror R, Criswell LA, Labetoulle M, Lietman TM, et al.; International Sjögren’s Syndrome Criteria Working Group. 2016 American College of Rheumatology/European League Against Rheumatism classification criteria for primary Sjögren’s syndrome: A consensus and data-driven methodology involving three international patient cohorts. Arthritis Rheumatol. 2017;69(1):35-45. doi:10.1002/art.39859; Petri M, Kim MY, Kalunian KC, Grossman J, Hahn BH, Sammaritano LR, et al.; OC-SELENA Trial. Combined oral contraceptives in women with systemic lupus erythematosus. N Engl J Med. 2005;353(24):2550-2558. doi:10.1056/NEJMoa051135; Seror R, Bowman SJ, Brito-Zeron P, Theander E, Bootsma H, Tzioufas A, et al. EULAR Sjögren’s syndrome disease activity index (ESSDAI): A user guide. RMD Open. 2015;1(1):e000022. doi:10.1136/rmdopen-2014-000022; Seror R, Ravaud P, Mariette X, Bootsma H, Theander E, Hansen A, et al.; EULAR Sjögren’s Task Force. EULAR Sjogren’s syndrome patient reported index (ESSPRI): Development of a consensus patient index for primary Sjogren’s syndrome. Ann Rheum Dis. 2011;70(6):968-972. doi:10.1136/ard.2010.143743; Зурочка АВ, Хайдуков СВ, Кудрявцев ИВ, Чернышев ВА. Проточная цитометрия в биомедицинских исследованиях. Екатеринбург:УрО РАН;2018.; Будкова АИ, Лапин СВ, Серебрякова МК, Кудрявцев ИВ, Тришина ИН, Маслянский АЛ, и др. Субпопуляционный состав В-клеток периферической крови у больных системной красной волчанкой. Медицинская иммунология. 2017;19(2):175-184.; Меснянкина АА, Соловьев СК, Александрова ЕН, Алексанкин АП, Супоницкая ЕВ, Елонаков АВ, и др. Динамика субпопуляции В-лимфоцитов у больных системной красной волчанкой на фоне терапии генно-инженерными биологическими препаратами. Научно-практическая ревматология. 2017;55(3):252-260.; Cornec D, Saraux A, Pers JO, Jousse-Joulin S, Marhadour T, Roguedas-Contios AM, et al. Diagnostic accuracy of blood B-cell subset profiling and autoimmunity markers in Sjögren’s syndrome. Arthritis Res Ther. 2014;16(1):R15. doi:10.1186/ar4442; Barcelos F, Martins C, Papoila A, Geraldes C, Cardigos J, Nunes G, et al. Association between memory B-cells and clinical and immunological features of primary Sjögren’s syndrome and Sicca patients. Rheumatol Int. 2018;38(6):1063-1073. doi:10.1007/s00296-018-4018-0; Ishioka-Takei E, Yoshimoto K, Suzuki K, Nishikawa A, Yasuoka H, Yamaoka K, et al. Increased proportion of a CD38highIgD+ B cell subset in peripheral blood is associated with clinical and immunological features in patients with primary Sjögren’s syndrome. Clin Immunol. 2018;187:85-91. doi:10.1016/j.clim.2017.10.008; Roberts ME, Kaminski D, Jenks SA, Maguire C, Ching K, Burbelo PD, et al. Primary Sjögren’s syndrome is characterized by distinct phenotypic and transcriptional profiles of IgD+ unswitched memory B cells. Arthritis Rheumatol. 2014;66(9):2558-2569. doi:10.1002/art.38734; Odendahl M, Jacobi A, Hansen A, Feist E, Hiepe F, Burmester GR, et al. Disturbed peripheral B lymphocyte homeostasis in systemic lupus erythematosus. J Immunol. 2000;165(10):5970-5979. doi:10.4049/jimmunol.165.10.5970; Hansen A, Odendahl M, Reiter K, Jacobi AM, Feist E, Scholze J, et al. Diminished peripheral blood memory B cells and accumulation of memory B cells in the salivary glands of patients with Sjögren’s syndrome. Arthritis Rheum. 2002;46(8):2160-2171. doi:10.1002/art.10445; Aqrawi LA, Brokstad KA, Jakobsen K, Jonsson R, Skarstein K. Low number of memory B cells in the salivary glands of patients with primary Sjögren’s syndrome. Autoimmunity. 2012;45(7):547-555. doi:10.3109/08916934.2012.712170; Mingueneau M, Boudaoud S, Haskett S, Reynolds TL, Nocturne G, Norton E, et al. Cytometry by time-of-flight immunophenotyping identifies a blood Sjögren’s signature correlating with disease activity and glandular inflammation. J Allergy Clin Immunol. 2016;137(6):1809-1821.e12. doi:10.1016/j.jaci.2016.01.024; Wardowska A, Komorniczak M, Skoniecka A, Bułło-Piontecka B, Lisowska KA, Dębska-Ślizień MA, et al. Alterations in peripheral blood B cells in systemic lupus erythematosus patients with renal insufficiency. Int Immunopharmacol. 2020;83:106451. doi:10.1016/j.intimp.2020.106451; Супоницкая ЕВ, Алексанкин АП, Меснянкина АА, Александрова ЕН, Панафидина ТА, Соловьев СК. Характеристика субпопуляций В-лимфоцитов периферической крови у больных активной системной красной волчанкой. Клиническая лабораторная диагностика. 2017;62(7):418-422.; Enocsson H, Karlsson J, Li HY, Wu Y, Kushner I, Wetterö J, et al. The complex role of C-reactive protein in systemic lupus erythematosus. J Clin Med. 2021;10(24):5837. doi:10.3390/jcm10245837

  3. 3
    Academic Journal

    المساهمون: Исследование выполнено при спонсорской поддержке АО «БИОКАД». Спонсор участвовал в разработке проекта исследования и поддержке исследовательской программы, а также в принятии решения о представлении статьи для публикации.

    المصدر: Rheumatology Science and Practice; Vol 62, No 1 (2024); 65-80 ; Научно-практическая ревматология; Vol 62, No 1 (2024); 65-80 ; 1995-4492 ; 1995-4484

    وصف الملف: application/pdf

    Relation: https://rsp.mediar-press.net/rsp/article/view/3514/2350; Эрдес ШФ, Бадокин ВВ, Бочкова АГ, Бугрова ОВ, Гайдукова ИЗ, Годзенко АА, и др. О терминологии спондилоартритов. Научно-практическая ревматология. 2015;53(6):657-660. doi:10.14412/1995-4484-2015-657-660; Baraliakos X, Kiltz U, Peters S, Appel H, Dybowski F, Igelmann M, et al. Efficiency of treatment with non-steroidal antiinflammatory drugs according to current recommendations in patients with radiographic and non-radiographic axial spondyloarthritis. Rheumatology (Oxford). 2017;56(1):95-102. doi:10.1093/rheumatology/kew367; Fragoulis GE, Siebert S. Treatment strategies in axial spondyloarthritis: What, when and how? Rheumatology (Oxford). 2020;59(Suppl 4):iv79-iv89.; Ramiro S, Nikiphorou E, Sepriano A, Ortolan A, Webers C, Baraliakos X, et al. ASAS-EULAR recommendations for the management of axial spondyloarthritis: 2022 update. Ann Rheum Dis. 2023;82(1):19-34.; Walsh JA, Saffore CD, Collins EB, Ostor A. Clinical and economic benefit of advanced therapies for the treatment of active ankylosing spondylitis. Rheumatol Ther. 2023;10(5):1385-1398.; Juanola X, Ramos MJM, Belzunegui JM, Fernández-Carballido C, Gratacós J. Treatment failure in axial spondyloarthritis: Insights for a standardized definition. Adv Ther. 2022;39(4): 1490-1501.; Deodhar A, Yu D. Switching tumor necrosis factor inhibitors in the treatment of axial spondyloarthritis. Semin Arthritis Rheum. 2017;47(3):343-350.; Del Vescovo S, Venerito V, Iannone C, Lopalco G. Uncovering the underworld of axial spondyloarthritis. Int J Mol Sci. 2023;24(7):6463. doi:10.3390/ijms24076463; Garrido-Mesa J, Brown MA. T cell repertoire profiling and the mechanism by which HLA-B27 causes ankylosing spondylitis. Curr Rheumatol Rep. 2022;24(12):398-410.; Komech EA, Pogorelyy MV, Egorov ES, Britanova OV, Rebrikov DV, Bochkova AG, et al. CD8+ T cells with characteristic T cell receptor beta motif are detected in blood and expanded in synovial fluid of ankylosing spondylitis patients. Rheumatology (Oxford). 2018;57(6):1097-1104.; Faham M, Carlton V, Moorhead M, Zheng J, Klinger M, Pepin F, et al. Discovery of T cell receptor β motifs specific to HLAB27-positive ankylosing spondylitis by deep repertoire sequence analysis. Arthritis Rheumatol. 2017;69(4):774-784.; Komech EA, Koltakova AD, Barinova AA, Minervina AA, Salnikova MA, Shmidt EI, et al. TCR repertoire profiling revealed antigen-driven CD8+ T cell clonal groups shared in synovial fluid of patients with spondyloarthritis. Front Immunol. 2022;13:973243.; Yang X, Garner LI, Zvyagin IV, Paley MA, Komech EA, Jude KM, et al. Autoimmunity-associated T cell receptors recognize HLA-B*27-bound peptides. Nature. 2022;612(7941):771-777. doi:10.1038/s41586-022-05501-7; Britanova OV, Lupyr KR, Staroverov DB, Shagina IA, Aleksandrov AA, Ustyugov YY, et al. Targeted depletion of TRBV9(+) T cells as immunotherapy in a patient with ankylosing spondylitis. Nat Med. 2023;29(11):2731-2736.; Израельсон МА, Степанов АВ, Староверов ДБ, Шагина ИВ, Мисорин АК, Евстратьева АВ, и др. Тестирование моноклональных антител к Т-клеточному рецептору, ассоциированному с анкилозирующим спондилитом. Вестник РГМУ. 2018;(5):83-92. doi:10.24075/brsmu.2018.064; European Medicines Agency. Guideline on the clinical investigation of medicinal products for the treatment of axial spondyloarthritis. EMA/CPMP/EWP/4891/03 Rev. 1, Corr. 1. 2017. URL: https://www.ema.europa.eu/en/clinical-investigationmedicinal-products-treatment-axial-spondyloarthritis#currenteffective-version-section (Accessed: 24th December 2021).; Sieper J, Rudwaleit M, Baraliakos X, Brandt J, Braun J, BurgosVargas R, et al. The Assessment of SpondyloArthritis international Society (ASAS) handbook: A guide to assess spondyloarthritis. Ann Rheum Dis. 2009;68(Suppl 2):ii1-ii44.; Maksymowych WP, Inman RD, Salonen D, Dhillon SS, Krishnananthan R, Stone M, et al. Spondyloarthritis Research Consortium of Canada magnetic resonance imaging index for assessment of spinal inflammation in ankylosing spondylitis. Arthritis Rheum. 2005;53(4):502-509.; van der Heijde D, Lie E, Kvien TK, Sieper J, van den Bosch F, Listing J, et al. ASDAS, a highly discriminatory ASAS-endorsed disease activity score in patients with ankylosing spondylitis. Ann Rheum Dis. 2009;68(12):1811-1818.; Гайдукова ИЗ, Ребров АП, Поддубный ДА. Эффективность и безопасность вводимого внутривенно метилпреднизолона для лечения пациентов с активным анкилозирующим спондилитом: результаты 12-недельного открытого пилотного исследования (МЕТАЛЛ). Терапевтический архив. 2015;87(5):47-52.; https://rsp.mediar-press.net/rsp/article/view/3514

  4. 4
    Academic Journal

    المساهمون: Статья подготовлена в рамках фундаментальной темы №1021051302580-4РК.

    المصدر: Modern Rheumatology Journal; Том 18, № 1 (2024); 47-53 ; Современная ревматология; Том 18, № 1 (2024); 47-53 ; 2310-158X ; 1996-7012

    وصف الملف: application/pdf

    Relation: https://mrj.ima-press.net/mrj/article/view/1530/1437; https://mrj.ima-press.net/mrj/article/view/1530/1448; Still GF. On a Form of Chronic Joint Disease in Children. Med Chir Trans. 1897; 80:47-60.9.; Bywaters EG. Still's disease in the adult. Ann Rheum Dis. 1971 Mar;30(2):121-33. doi:10.1136/ard.30.2.121.; Nirmala N, Brachat A, Feist E, et al. Geneexpression analysis of adult-onset Still`s disease and systemic juvenile idiopathic arthritis is consistent with a continuum of a 46 single disease entity. Pediatr Rheumatol Online J. 2015 Nov 20;13:50. doi:10.1186/s12969-015-0047-3; Yamaguchi M, Ohta A, Tsunematsu T, et al. Preliminary criteria for classification of adult Still's disease. J Rheumatol. 1992 Mar;19(3): 424-30.; Насонов ЕЛ, Файст Е. Болезнь Стилла взрослых: новые горизонты. Научно-практическая ревматология. 2021;59(6): 645-665.; Pouchot J, Sampalis JS, Beaudet F, et al. Adult Still's disease: manifestations, disease course, and outcome in 62 patients. Medicine (Baltimore). 1991 Mar;70(2):118-36.; Ruscitti P, Cipriani P, Masedu F, et al. Adult-onset Still's disease: evaluation of prognostic tools and validation of the systemic score by analysis of 100 cases from three centers. BMC Med. 2016 Dec 1;14(1):194. doi:10.1186/s12916-016-0738-8.; Ichida H, Kawaguchi Y, Sugiura T, et al. Clinical manifestations of Adult-onset Still's disease presenting with erosive arthritis: Association with low levels of ferritin and Interleukin-18. Arthritis Care Res (Hoboken). 2014 Apr;66(4):642-6. doi:10.1002/acr.22194.; Maria AT, Le Quellec A, Jorgensen C, et al. Adult onset Still's disease (AOSD) in the era of biologic therapies: dichotomous view for cytokine and clinical expressions. Autoimmun Rev. 2014 Nov;13(11):1149-59. doi:10.1016/j.autrev.2014.08.032.; Mitrovic S, Fautrel B. New Markers for Adult-Onset Still s Disease. Joint Bone Spine. 2018 May;85(3):285-293. doi:10.1016/j.jbspin. 2017.05.011.; Wouters JM, van de Putte LB. Adult-onset Still's disease; clinical and laboratory features, treatment and progress of 45 cases. Q J Med. 1986 Nov;61(235):1055-65.; Fautrel B, Borget C, Rozenberg S, et al. Corticosteroid sparing effect of low dose methotrexate treatment in adult Still's disease. J Rheumatol. 1999 Feb;26(2):373-8.; Ravelli A, Ramenghi B, Di Fuccia G, et al. Factors associated with response to methotrexate in systemic-onset juvenile chronic arthritis. Acta Paediatr. 1994 Apr;83(4):428-32. doi:10.1111/j.1651-2227.1994.tb18135.x.; Woo P, Southwood TR, Prieur AM, et al. Randomized, placebo-controlled, crossover trial of low-dose oral methotrexate in children with extended oligoarticular or systemic arthritis. Arthritis Rheum. 2000 Aug;43(8): 1849-57. doi:10.1002/1529-0131(200008)43:83.0.CO;2-F.; Tomaras S, Goetzke CC, Kallinich T, et al. Adult-Onset Still’s Disease: Clinical Aspects and Therapeutic Approach. J Clin Med. 2021 Feb 12;10(4):733. doi:10.3390/jcm10040733.; Myachikova V, Moiseeva O, Konradi A, et al. A retrospective analysis of colchicine in combination with NSAIDs therapy in patients with systemic form of adult-onset Still disease with serositis. Clin Exp Rheumatol. 2022 Sep; 40(8):1474-1479. doi:10.55563/clinexprheumatol/1o41c8.; Asano T, Furuya MY, Sato S, et al. Adding colchicine to immunosuppressive treatments; a potential option for biologics-refractory adult-onset Still's disease. BMC Res Notes. 2018 May 21;11(1):320. doi:10.1186/s13104-018-3420-8.; Ou-Yang LJ, Tang KT. A case of adult onset Still's disease with mutations of the MEFV gene who is partially responsive to colchicine. Medicine (Baltimore). 2018 Apr; 97(15):e0333. doi:10.1097/MD.0000000000010333.; Rao S, Shi W. A case of adult-onset Still's disease accompanied with pulmonary tuberculosis successfully treated with colchicine. Postepy Dermatol Alergol. 2021 Oct;38(5):912- 915. doi:10.5114/ada.2021.110105.; Мячикова ВЮ, Маслянский АЛ, Гайдукова ИЗ и др. Трудности диагностики и лечения болезни Стилла взрослых, протекавшей с экссудативным перикардитом в качестве ведущего клинического проявления. Современная ревматология. 2016; 10(1):31-36. doi:10.14412/1996-7012-2016-1-31-36.; Mehta BY, Ibrahim S, Briggs W, Efthimiou P. Racial/Ethnic variations in morbidity and mortality in Adult Onset Still's Disease: An analysis of national dataset. Semin Arthritis Rheum. 2019 Dec;49(3):469-473. doi:10.1016/j.semarthrit.2019.04.004.; Sakata N, Shimizu S, Hirano F, Fushimi K. Epidemiological study of adult-onset Still's disease using a Japanese administrative database. Rheumatol Int. 2016 Oct;36(10): 1399-405. doi:10.1007/s00296-016-3546-8.; https://mrj.ima-press.net/mrj/article/view/1530

  5. 5
    Academic Journal

    المساهمون: Исследование выполнено при финансовой поддержке Министерства науки и высшего образования Российской Федерации (соглашение № 075-15-2022-301 от 20.04.2022).

    المصدر: Rheumatology Science and Practice; Vol 62, No 1 (2024); 90-97 ; Научно-практическая ревматология; Vol 62, No 1 (2024); 90-97 ; 1995-4492 ; 1995-4484

    وصف الملف: application/pdf

    Relation: https://rsp.mediar-press.net/rsp/article/view/3517/2352; Насонов ЕЛ (ред.). Ревматология. Российские клинические рекомендации. М.:ГЭОТАР-Медиа;2017.; Насонов ЕЛ. Перспективы анти-В-клеточной терапии в ревматологии. Научно-практическая ревматология. 2018;56(5):539-548. doi:10.14412/1995-4484-2018-539-548; Шадуро ДВ, Белоглазов ВА, Гордиенко АИ. Уровни основных субпопуляций лимфоцитов и их связь с клеточным и гуморальным звеном антиэндотоксинового иммунитета у больных системной красной волчанкой. Медицинская иммунология. 2015;17(4):359-366. doi:10.15789/1563-0625-2015-4-359-366; Blanco P, Pitard V, Viallard JF, Taupin JL, Pellegrin JL, Moreau JF. Increase in activated CD8+ T lymphocytes expressing perforin and granzyme B correlates with disease activity in patients with systemic lupus erythematosus. Arthritis Rheum. 2005;52:201-211. doi:10.1002/art.20745; Couzi L, Merville P, Deminière C, Moreau JF, Combe C, Pellegrin JL, et al. Predominance of CD8+ T lymphocytes among periglomerular infiltrating cells and link to the prognosis of class III and class IV lupus nephritis. Arthritis Rheum. 2007;56(7):2362-2370. doi:10.1002/art.22654; Dolff S, Abdulahad WH, Arends S, van Dijk MC, Limburg PC, Kallenberg CG, et al. Urinary CD8+ T-cell counts discriminate between active and inactive lupus nephritis. Arthritis Res Ther. 2013;15(1):R36. doi:10.1186/ar4189; Klocke J, Kopetschke K, Grießbach AS, Langhans V, Humrich JY, Biesen R, et al. Mapping urinary chemokines in human lupus nephritis: Potentially redundant pathways recruit CD4+ and CD8+ T cells and macrophages. Eur J Immunol. 2017;47(1):180-192. doi:10.1002/eji.201646387; Yuan S, Zeng Y, Li J, Wang C, Li W, He Z, et al. Phenotypical changes and clinical significance of CD4+/CD8+ T cells in SLE. Lupus Sci Med. 2022;9(1):e000660. doi:10.1136/lupus-2022-000660; St Paul M, Ohashi PS. The roles of CD8+ T cell subsets in antitumor immunity. Trends Cell Biol. 2020;30(9):695-704. doi:10.1016/j.tcb.2020.06.003; Fousteri G, Kuka M. The elusive identity of CXCR5+ CD8 T cells in viral infection and autoimmunity: Cytotoxic, regulatory, or helper cells? Mol Immunol. 2020;119:101-105. doi:10.1016/j.molimm.2020.01.007; Aringer M, Costenbader K, Daikh D, Brinks R, Mosca M, Ramsey-Goldman R, et al. 2019 European League Against Rheumatism/American College of Rheumatology classification criteria for systemic lupus erythematosus. Arthritis Rheumatol. 2019;71(9):1400-1412. doi:10.1002/art.40930; Кудрявцев ИВ, Борисов АГ, Кробинец ИИ, Савченко АА, Серебрякова МК, Тотолян АА. Хемокиновые рецепторы на Т-хелперах различного уровня дифференцировки: основные субпопуляции. Медицинская иммунология. 2016;18(3):239-250. doi:10.15789/1563-0625-2016-3-239-250; Loyal L, Warth S, Jürchott K, Mölder F, Nikolaou C, Babel N, et al. SLAMF7 and IL-6R define distinct cytotoxic versus helper memory CD8+ T cells. Nat Commun. 2020;11(1):6357. doi:10.1038/s41467-020-19002-6; Kudryavtsev IV, Arsentieva NA, Korobova ZR, Isakov DV, Rubinstein AA, Batsunov OK, et al. Heterogenous CD8+ T cell maturation and ‘polarization’ in acute and convalescent COVID19 patients. Viruses. 2022;14(9):1906. doi:10.3390/v14091906; Chen J, Ding L, Meng W, Yang J, Yan C, Xie J, et al. Vincristinecyclophosphamide combination therapy positively affects T-cell subset distribution in systemic lupus erythematosus patients. Med Sci Monit. 2015;21:505-510. doi:10.12659/MSM.893271; Chen PM, Tsokos GC. The role of CD8+ T-cell systemic lupus erythematosus pathogenesis: An update. Curr Opin Rheumatol. 2021;33(6):586-591. doi:10.1097/BOR.0000000000000815; Postal M, Appenzeller S. The role of tumor necrosis factor-alpha (TNF-α) in the pathogenesis of systemic lupus erythematosus. Cytokine. 2011;56(3):537-543. doi:10.1016/j.cyto.2011.08.026; Jacob CO, McDevitt HO. Tumor necrosis factor-alpha in murine autoimmune ‘lupus’ nephritis. Nature. 1988;331:356-358. doi:10.1038/331356a0; López P, Gutiérrez C, Suárez A. IL-10 and TNFα genotypes in SLE. J Biomed Biotechnol. 2010;2010:838390. doi:10.1155/2010/838390; Bridgewood C, Wittmann M, Macleod T, Watad A, Newton D, Bhan K, et al. T helper 2 IL-4/IL-13 dual blockade with dupilumab is linked to some emergent T helper 17-type diseases, including seronegative arthritis and enthesitis/enthesopathy, but not to humoral autoimmune diseases. J Invest Dermatol. 2022;142(10):2660-2667. doi:10.1016/j.jid.2022.03.013; https://rsp.mediar-press.net/rsp/article/view/3517

  6. 6
    Academic Journal

    المصدر: Modern Rheumatology Journal; Том 17, № 1 (2023); 24–30 ; Современная ревматология; Том 17, № 1 (2023); 24–30 ; 2310-158X ; 1996-7012

    وصف الملف: application/pdf

    Relation: https://mrj.ima-press.net/mrj/article/view/1384/1322; Shiboski CH, Shiboski SC, Seror R, et al. 2016 American College of Rheumatology/European League Against Rheumatism Classification Criteria for Primary Sjцgren's Syndrome: A Consensus and Data-Driven Methodology Involving Three International Patient Cohorts. Arthritis Rheumatol. 2017 Jan;69(1): 35-45. doi:10.1002/art.39859. Epub 2016 Oct 26.; Qin B, Wang J, Yang Z, et al. Epidemiology of primary Sjцgren's syndrome: a systematic review and meta-analysis. Ann Rheum Dis. 2015 Nov;74(11):1983-9. doi:10.1136/annrheumdis-2014-2053. Epub 2014 Jun 17.; Caporali R, Bonacci E, Epis O, et al. Safety and usefulness of minor salivary gland biopsy: retrospective analysis of 502 procedures performed at a single center. Arthritis Rheum. 2008 May 15;59(5):714-20. doi:10.1002/art.23579.; Thorlacius GE, Hultin-Rosenberg L, Sandling JK, et al. Genetic and clinical basis for two distinct subtypes of primary Sjоgren's syndrome. Rheumatology (Oxford). 2021 Feb 1; 60(2):837-48. doi:10.1093/rheumatology/keaa367.; Carubbi F, Alunno A, Cipriani P, et al. Different operators and histologic techniques in the assessment of germinal center-like structures in primary Sjцgren's syndrome minor salivary glands. PLoS One. 2019 Jan 25;14(1):e0211142. doi:10.1371/journal.pone.0211142. eCollection 2019.; Shoenfeld Y, Meroni PL, Gershwin ME. Autoantibodies. Waltham: Elsevier; 2014. P. 233-8.; Brito-Zerуn P, Acar-Denizli N, Zeher M, et al. How does primary Sjögren syndrome present in biopsy-proven patients without circulating rola autoantibodies? Characteristics at diagnosis of 2073 patients from the Sjögren big data project. https://acrabstracts.org/abstract/sjogren-big-data-project-influence-ofgeolocation-on-the-phenotypic-expressionat-diagnosis-in-8310-patients-north-tosouth-gradient/; Aqrawi LA, Galtung HK, Guerreiro EM, et al. Proteomic and histopathological characterisation of sicca subjects and primary Sjögren's syndrome patients reveals promising tear, saliva and extracellular vesicle disease biomarkers. Arthritis Res Ther. 2019 Jul 31; 21(1):181. doi:10.1186/s13075-019-1961-4.; Christodoulou MI, Kapsogeorgou EK, Moutsopoulos HM. Characteristics of the minor salivary gland infiltrates in Sjögren's syndrome. J Autoimmun. 2010 Jun;34(4):400-7. doi:10.1016/j.jaut.2009.10.004. Epub 2009 Nov 3.; Nocturne G, Mariette X. B cells in the pathogenesis of primary Sjögren syndrome. Nat Rev Rheumatol. 2018 Mar;14(3):133-45. doi:10.1038/nrrheum.2018.1. Epub 2018 Feb 8.; Sandhya P, Christudoss P, Kabeerdoss J, et al. Diagnostic accuracy of salivary and serum-free light chain assays in primary Sjögren's syndrome: a pilot study. Int J Rheum Dis. 2017 Jun;20(6):760-766. doi:10.1111/1756-185X.12965. Epub 2016 Dec 30.; Verstappen GM, Moerman RV, van Nimwegen JF, et al. Serum immunoglobulin free light chains are sensitive biomarkers for monitoring disease activity and treatment response in primary Sjögren's syndrome. Rheumatology (Oxford). 2018 Oct 1;57(10):1812-21. doi:10.1093/rheumatology/key180.; Navazesh M, Kumar SK. Measuring salivary flow: challenges and opportunities. J Am Dent Assoc. 2008 May;139 Suppl:35S-40S. doi:10.14219/jada.archive.2008.; Тотолян АА. Современные подходы к диагностике иммунопатологических состояний. Медицинская иммунология. 1999;1(1-2):75-108.; Teppo H, Revonta M. A follow-up study of minimally invasive lip biopsy in the diagnosis of Sjögren's syndrome. Clin Rheumatol. 2007 Jul;26(7):1099-103. doi:10.1007/s10067-006-0457-1. Epub 2006 Nov 25.; Katsiougiannis S, Wong DTW. The Proteomics of Saliva in Sjögren's Syndrome. Rheum Dis Clin North Am. 2016;42(3):449-56. doi:10.1016/j.rdc.2016.03.004. Epub 2016 Jun 21.; Szyszko EA, Brokstad KA, Oijordsbakken G, et al. Salivary glands of primary Sjögren's syndrome patients express factors vital for plasma cell survival. Arthritis Res Ther. 2011 Jan 7;13(1):R2. doi:10.1186/ar3220.; Mingueneau M, Boudaoud S, Haskett S, et al. Cytometry by time-of-flight immunophenotyping identifies a blood Sjögren's signature correlating with disease activity and glandular inflammation. J Allergy Clin Immunol. 2016 Jun;137(6):1809-21.e12. doi:10.1016/j.jaci.2016.01.024. Epub 2016 Apr 1.; Будкова АИ, Лапин СВ, Серебрякова МК и др. Субпопуляционный состав В-клеток периферической крови у больных системной красной волчанкой. Медицинская иммунология. 2017;19(2):175-84.; Hamza N, Bootsma H, Yuvaraj S, et al. Persistence of immunoglobulin-producing cells in parotid salivary glands of patients with primary Sjögren's syndrome after B cell depletion therapy. Ann Rheum Dis. 2012 Nov;71(11): 1881-7. doi:10.1136/annrheumdis-2011-201189. Epub 2012 May 21.; Takeshita M, Suzuki K, Kaneda Y, et al. Antigen-driven selection of antibodies against SSA, SSB and the centromere 'complex', including a novel antigen, MIS12 complex, in human salivary glands. Ann Rheum Dis. 2020 Jan;79(1):150-8. doi:10.1136/annrheumdis-2019-215862. Epub 2019 Oct 14.; Bookman AA, Shen H, Cook RJ, et al. Whole stimulated salivary flow: correlation with the pathology of inflammation and damage in minor salivary gland biopsy specimens from patients with primary Sjögren's syndrome but not patients with sicca. Arthritis Rheum. 2011 Jul;63(7):2014-20. doi:10.1002/art.30295.; Tarn JR, Howard-Tripp N, Lendrem DW, et al. Symptom-based stratification of patients with primary Sjögren's syndrome: multi-dimensional characterisation of international observational cohorts and reanalyses of randomised clinical trials. Lancet Rheumatol. 2019 Sep 25;1(2):e85-94. doi:10.1016/S2665-9913(19)30042-6.; Sandhya P, Kabeerdoss J, Christudoss P, et al. Salivary free light chains and salivary immunoglobulins as potential non-invasive biomarkers in primary Sjögren's syndrome. Int J Rheum Dis. 2022;25(1):61-9. doi:10.1111/1756-185X.14242. Epub 2021 Nov 17.; Fox PC, Datiles M, Atkinson JC, et al. Prednisone and piroxicam for treatment of primary Sjögren's syndrome. Clin Exp Rheumatol. 1993 Mar-Apr;11(2):149-56.; https://mrj.ima-press.net/mrj/article/view/1384

  7. 7
    Academic Journal

    المصدر: Rheumatology Science and Practice; Vol 60, No 5 (2022); 603-608 ; Научно-практическая ревматология; Vol 60, No 5 (2022); 603-608 ; 1995-4492 ; 1995-4484

    وصف الملف: application/pdf

    Relation: https://rsp.mediar-press.net/rsp/article/view/3230/2234; van den Hoogen F, Khanna D, Fransen J, et al. 2013 classification criteria for systemic sclerosis: an American college of rheumatology/European league against rheumatism collaborative initiative. Ann Rheum Dis. 2013;72(11):1747–1755. doi:10.1136/annrheumdis-2013-204424; Park EH, Strand V, Oh YJ, Song YW, Lee EB. Health-related quality of life in systemic sclerosis compared with other rheumatic diseases: a cross-sectional study. Arthritis Res Ther. 2019;21(1):61. Published 2019 Feb 15. doi:10.1186/s13075-019-1842-x; van Leeuwen NM, Ciaffi J, Liem SIE, et al. Health-related quality of life in patients with systemic sclerosis: evolution over time and main determinants. Rheumatology (Oxford). 2021;60(8):3646– 3655. doi:10.1093/rheumatology/keaa827; Denton CP, Khanna D. Systemic sclerosis. Lancet. 2017; 390(10103):1685–1699. doi:10.1016/S0140-6736(17)30933-9; Yen EY, Singh DR, Singh RR. Trends in Systemic Sclerosis Mortality Over Forty-Eight Years, 1968–2015: A US Population-Based Study. Arthritis Care Res (Hoboken). 2021;73(10):1502–1510. doi:10.1002/acr.24411; Kowal-Bielecka O, Fransen J, Avouac J, et al. Update of EULAR recommendations for the treatment of systemic sclerosis. Ann Rheum Dis. 2017;76(8):1327–1339. doi:10.1136/annrheumdis-2016-209909; Naidu GSRSNK, Sharma SK, Adarsh MB, et al. Effect of mycophenolate mofetil (MMF) on systemic sclerosis-related interstitial lung disease with mildly impaired lung function: a doubleblind, placebo-controlled, randomized trial. Rheumatol Int. 2020;40(2):207–216. doi:10.1007/s00296-019-04481-8; Ананьева Л.П. Современная терапия интерстициальных пневмоний, ассоциированных с системной склеродермией. Научно-практическая ревматология. 2020;58(5):520–531.; Bruni T, Varone F. The adoption of nintedanib in systemic sclerosis: the SENSCIS study. Breathe (Sheff). 2020;16(2):200005. doi:10.1183/20734735.0005-2020; de Figueiredo Caldas MMV, de Azevedo KPM, de França Nunes AC, et al. Is rituximab effective for systemic sclerosis? A systematic review and meta-analysis. Adv Rheumatol. 2021;61(1):15. Published 2021 Feb 27. doi:10.1186/s42358-021-00170-y; Khanna D, Denton CP, Jahreis A, et al. Safety and efficacy of subcutaneous tocilizumab in adults with systemic sclerosis (faSScinate): a phase 2, randomised, controlled trial [published correction appears in Lancet. 2018 Apr 7;391(10128):1356]. Lancet. 2016;387(10038):2630–2640. doi:10.1016/S0140-6736(16)00232-4; Khanna D, Lin CJF, Furst DE, et al. Tocilizumab in systemic sclerosis: a randomised, double-blind, placebo-controlled, phase 3 trial [published correction appears in Lancet Respir Med. 2020 Oct;8(10):e75] [published correction appears in Lancet Respir Med. 2021 Mar;9(3):e29]. Lancet Respir Med. 2020;8(10):963–974. doi:10.1016/S2213-2600(20)30318-0; Невская Т.А., Новиков А.А., Александрова Е.Н., Мач Э.С., Запрягаева М.Н., Сперанский А.И., Гусева Н.Г., Ананьева Л.П. Клиническое значение высокочувствительного С-реактивного белка при системной склеродермии. Научно-практическая ревматология. 2007;45(4):10–17.; Cardonea nu A, Burlui AM, Macovei LA, et al. Targeting Systemic Sclerosis from Pathogenic Mechanisms to Clinical Manifestations: Why IL-6?. Biomedicines. 2022;10(2):318. Published 2022 Jan 29. doi:10.3390/biomedicines10020318; Ананьева ЛП. Перспективы применения тоцилизумаба при системной склеродермии. Научно-практическая ревматология. 2015;53(6):632–640.; Avci AB, Feist E, Burmester GR. Targeting IL-6 or IL-6 Receptor in Rheumatoid Arthritis: What’s the Difference?. BioDrugs. 2018;32(6):531–546. doi:10.1007/s40259-018-0320-3; Hosoya H, Derk CT. Clinically Symptomatic Pericardial Effusions in Hospitalized Systemic Sclerosis Patients: Demographics and Management. Biomed Res Int. 2018;2018:6812082. Published 2018 Jun 4. doi:10.1155/2018/6812082; Ufuk F, Çakmak P, Sağtaş E, et al. Extra-parenchymal chest HRCT findings of patients with systemic sclerosis at the time of initial diagnosis. Bezmialem Science. 2018;7(2):101–106. doi:10.14235/bs.2018.2427; Ocampo V, Haaland D, Legault K, et al. Successful treatment of recurrent pleural and pericardial effusions with tocilizumab in a pa - tient with systemic lupus erythematous. BMJ Case Rep. 2016;2016: bcr2016215423. Published 2016 Aug 8. doi:10.1136/bcr-2016-215423; Yoshida S, Takeuchi T, Sawaki H, et al. Successful treatment with tocilizumab of pericarditis associated with rheumatoid arthritis. Mod Rheumatol. 2014;24(4):677–680. doi:10.3109/14397595.2013.874733; Zacay G, Levy Y. Outcomes of patients with systemic sclerosis treated with tocilizumab: Case series and review of the literature. Best Pract Res Clin Rheumatol. 2018;32(4):563–571. doi:10.1016/j.berh.2019.01.011; Kawaguchi Y. Contribution of Interleukin-6 to the Pathogenesis of Systemic Sclerosis. Journal of Scleroderma and Related Disorders. 2017;2(2):6–12. doi:10.5301/jsrd.5000258; Hernández-Sánchez J, Harlow L, Church C, et al. Clinical trial protocol for TRANSFORM-UK: A therapeutic open-label study of tocilizumab in the treatment of pulmonary arterial hyperten - sion. Pulm Circ. 2018;8(1):2045893217735820. doi:10.1177/2045893217735820; https://rsp.mediar-press.net/rsp/article/view/3230

  8. 8
    Academic Journal

    المساهمون: Исследование выполнялось в рамках гранта Министерства науки и высшего образования Российской Федерации (договор № 075-15-2020-901).

    المصدر: Rheumatology Science and Practice; Vol 60, No 3 (2022); 341-346 ; Научно-практическая ревматология; Vol 60, No 3 (2022); 341-346 ; 1995-4492 ; 1995-4484

    وصف الملف: application/pdf

    Relation: https://rsp.mediar-press.net/rsp/article/view/3182/2196; Feist E, Mitrovic S, Fautrel B. Mechanisms, biomarkers and targets for adult-onset Still’s disease. Nat Rev Rheumatol. 2018;14(10):603-618. doi:10.1038/s41584-018-0081-x; Hung WT, Chen YM, Hung SI, Chen HH, Gung NR, Hsieh CW, et al. CARD8 SNP rs11672725 identified as a potential genetic variant for adult-onset Still’s disease. Life (Basel). 2021;11(5):382. doi:10.3390/life11050382 3. The portal for rare diseases and orphan drugs. URL: https://www.orpha.net/consor/cgi-bin/Disease_Search.php?lng=EN&data_id=5525&Disease_Disease_Search_diseaseGroup=AOSD&Disease_Disease_Search_diseaseType (последнее обновление 2019г); Cush JJ, Medsger TA Jr, Christy WC, Herbert DC, Cooperstein LA. Adult-onset Still’s disease. Clinical course and outcome. Arthritis Rheum. 1987;30(2):186-194. doi:10.1002/art.1780300209; Yamaguchi M, Ohta A, Tsunematsu T, Kasukawa R, Mizushima Y, Kashiwagi H, et al. Preliminary criteria for classification of adult Still’s disease. J Rheumatol. 1992;19(3):424-430.; Pepys MB, Hirschfield GM. C-reactive protein: A critical update. J Clin Invest. 2003;111(12):1805-1812. doi:10.1172/JCI18921; Riley LK, Rupert J. Evaluation of patients with leukocytosis. Am Fam Physician. 2015;92(11):1004-1011.; Wang W, Knovich MA, Coffman LG, Torti FM, Torti SV. Serum ferritin: Past, present and future. Biochim Biophys Acta. 2010;1800(8):760-769. doi:10.1016/j.bbagen.2010.03.011; Zandman-Goddard G, Shoenfeld Y. Ferritin in autoimmune diseases. Autoimmun Rev. 2007;6(7):457-463. doi:10.1016/j.autrev.2007.01.016; Ozen S, Demirkaya E, Erer B, Livneh A, Ben-Chetrit E, Giancane G, et al. EULAR recommendations for the management of familial Mediterranean fever. Ann Rheum Dis. 2016;75(4):644- 651. doi:10.1136/annrheumdis-2015-208690; Averill MM, Barnhart S, Becker L, Li X, Heinecke JW, Leboeuf RC, et al. S100A9 differentially modifies phenotypic states of neutrophils, macrophages, and dendritic cells: Implications for atherosclerosis and adipose tissue inflammation. Circulation. 2011;123(11):1216- 1226. doi:10.1161/CIRCULATIONAHA.110.985523; Mariani A, Marsili M, Nozzi M, Faricelli R, Chiarelli F, Breda L. Serum calprotectin: Review of its usefulness and validity in paediatric rheumatic diseases. Clin Exp Rheumatol. 2015;33(1):109-114.; Авдеева АС. Клиническое значение кальпротектина при ревматических заболеваниях. Научно-практическая ревматология. 2018;56(4):494-499. doi:10.14412/1995-4484-2018-494-499; Colafrancesco S, Priori R, Alessandri C, Perricone C, Pendolino M, Picarelli G, et al. IL-18 serum level in adult onset Still’s disease: A marker of disease activity. Int J Inflam. 2012;2012:156890. doi:10.1155/2012/156890; Мячикова ВЮ, Маслянский АЛ, Ткаченко ОЮ, Первакова МЮ, Кувардин ЕС, Лапин СВ. Сравнительный анализ концентраций провоспалительных цитокинов и гликозилированного ферритина у пациентов с идиопатическим рецидивирующим перикардитом и болезнью Стилла взрослых. Российский кардиологический журнал. 2021;26(11):4715. doi:10.15829/1560-4071-2021-4715; Лапин СВ, Маслянский АЛ, Лазарева НМ, Васильева ЕЮ, Тотолян АА. Значение количественного определения прокальцитонина для диагностики септических осложнений у больных с аутоиммунными ревматическими заболеваниями. Клиническая лабораторная диагностика. 2013;1:28-33.; Hamade B, Huang DT. Procalcitonin: Where are we now? Crit Care Clin. 2020;36(1):23-40. doi:10.1016/j.ccc.2019.08.003; Буханова ДВ, Белов БС, Тарасова ГМ, Дилбарян АГ. Прокальцитониновый тест в ревматологии. Клиницист. 2017;11(2): 16-23. doi:10.17650/1818-8338-2017-11-2-16-23; van Riel PL, Renskers L. The Disease Activity Score (DAS) and the Disease Activity Score using 28 joint counts (DAS28) in the management of rheumatoid arthritis. Clin Exp Rheumatol. 2016;34(5 Suppl 101):S40-S44.; Kedor C, Listing J, Zernicke J, Weiß A, Behrens F, Blank N, et al. Canakinumab for Treatment of Adult-Onset Still’s Disease to Achieve Reduction of Arthritic Manifestation (CONSIDER): Phase II, randomised, double-blind, placebo-controlled, multicentre, investigator-initiated trial. Ann Rheum Dis. 2020;79(8):1090-1097. doi:10.1136/annrheumdis-2020-217155; Pouchot J, Sampalis JS, Beaudet F, Carette S, Décary F, Salusinsky-Sternbach M, et al. Adult Still’s disease: Manifestations, disease course, and outcome in 62 patients. Medicine (Baltimore). 1991;70(2):118-136.; Ruscitti P, Cipriani P, Masedu F, Iacono D, Ciccia F, Liakouli V, et al. Adult-onset Still’s disease: Evaluation of prognostic tools and validation of the systemic score by analysis of 100 cases from three centers. BMC Med. 2016;14(1):194. doi:10.1186/s12916-016-0738-8; Насонов ЕЛ, Файст Е. Болезнь Стилла взрослых: новые горизонты. Научно-практическая ревматология. 2021;59(6):645- 665. doi:10.47360/1995-4484-2021-643-663; Потапенко ВГ, Первакова МЮ, Лапин СВ, Титов АК, Суркова ЕА, Петрова НН, и др. Роль фракционного анализа ферритина в диагностике вторичного гемофагоцитарного синдрома. Клиническая лабораторная диагностика. 2018;63(1):21-27. doi:10.18821/0869-2084-2018-63-1-21-27; United Nations, Department of Economic and Social Affairs, Population Division. World population ageing 2015 (ST/ESA/ SER.A/390). 2015. 26. Dinarello CA. Overview of the IL-1 family in innate inflammation and acquired immunity. Immunol Rev. 2018;281(1):8-27. doi:10.1111/imr.12621; Priori R, Colafrancesco S, Alessandri C, Minniti A, Perricone C, Iaiani G, et al. Interleukin 18: A biomarker for differential diagnosis between adult-onset Still’s disease and sepsis. J Rheumatol. 2014;41(6):1118-1123. doi:10.3899/jrheum.130575; Rau M, Schiller M, Krienke S, Heyder P, Lorenz H, Blank N. Clinical manifestations but not cytokine profiles differentiate adult-onset Still’s disease and sepsis. J Rheumatol. 2010;37(11):2369-2376. doi:10.3899/jrheum.100247; Scheinberg MA, Chapira E, Fernandes ML, Hubscher O. Interleukin 6: A possible marker of disease activity in adult onset Still’s disease. Clin Exp Rheumatol. 1996;14(6):653-655.; Zhang W, Yang T, Zhang H, Xu Y, Yang Q, Liu Q, et al. Biomarker screening and validation for the differentiation of bloodstream infection from adult-onset Still’s disease: A prospective cohort study. Cytokine. 2021;146:155642. doi:10.1016/j.cyto.2021.155642; https://rsp.mediar-press.net/rsp/article/view/3182

  9. 9
    Academic Journal

    المصدر: Rheumatology Science and Practice; Vol 60, No 3 (2022); 381-387 ; Научно-практическая ревматология; Vol 60, No 3 (2022); 381-387 ; 1995-4492 ; 1995-4484

    وصف الملف: application/pdf

    Relation: https://rsp.mediar-press.net/rsp/article/view/3188/2202; Steen VD, Medsger TA. Changes in causes of death in systemic sclerosis, 1972–2002. Ann Rheum Dis. 2007;66(7):940-944. doi:10.1136/ard.2006.066068; Goldin JG, Kim GHJ, Tseng CH, Volkmann E, Furst D, Clements P, et al. Longitudinal changes in quantitative interstitial lung disease on computed tomography after immunosuppression in the Scleroderma Lung Study II. Ann Am Thorac Soc. 2018;15(11):1286-1295. doi:10.1513/AnnalsATS.201802-079OC; Giuggioli D, Lumetti F, Colaci M, Fallahi P, Antonelli A, Ferri C. Rituximab in the treatment of patients with systemic sclerosis. Our experience and review of the literature. Autoimmun Rev. 2015;14(11):1072-1078. doi:10.1016/j.autrev.2015.07.008; Zacay G, Levy Y. Outcomes of patients with systemic sclerosis treated with tocilizumab: Case series and review of the literature. Best Pract Res Clin Rheumatol. 2018;32(4):563-571. doi:10.1016/j.berh.2019.01.011; Авдеев СН, Ананьева ЛП, Жиляев ЕВ, Зонова ЕВ, Клименко АА, Конева ОА, и др. Резолюция Совета экспертов, посвященного теме «Интерстициальные заболевания легких при системной склеродермии» (14 октября 2019 г., Москва). Современная ревматология. 2020;14(1):125-128. doi:10.14412/1996-7012-2020-1-125-128; Новик АА, Богданов АН. Принципы трансплантации костного мозга и стволовых клеток периферической крови. СПб.:Военно-медицинская академия;2001.; European Society for Blood and Marrow Transplantation. Transplant activity survey. URL: https://www.ebmt.org/registry/transplant-activity-survey (Accessed: DD Month 2022).; Burt RK, Shah SJ, Dill K, Grant T, Gheorghiade M, Schroeder J, et al. Autologous non-myeloablative haemopoietic stem-cell transplantation compared with pulse cyclophosphamide once per month for systemic sclerosis (ASSIST): An open-label, randomised phase 2 trial. Lancet. 2011;378(9790):498-506. doi:10.1016/S0140-6736(11)60982-3; van Laar JM, Farge D, Sont JK, Naraghi K, Marjanovic Z, Larghero J, et al.; EBMT/EULAR Scleroderma Study Group. Autologous hematopoietic stem cell transplantation vs intravenous pulse cyclophosphamide in diffuse cutaneous systemic sclerosis: A randomized clinical trial. JAMA. 2014;311(24):2490-2498. doi:10.1001/jama.2014.6368; Sullivan KM, Goldmuntz EA, Keyes-Elstein L, McSweeney PA, Pinckney A, Welch B, et al.; SCOT Study Investigators. Myeloablative autologous stem-cell transplantation for severe scleroderma. N Engl J Med. 2018;378(1):35-47. doi:10.1056/nejmoa1703327; Farge D. Stem cell transplantation for systemic sclerosis in Israel: A new star is rising. Isr Med Assoc J. 2020;22(2):116-118.; Kowal-Bielecka O, Fransen J, Avouac J, Becker M, Kulak A, Allanore Y, et al.; EUSTAR Coauthors. Update of EULAR recommendations for the treatment of systemic sclerosis. Ann Rheum Dis. 2017;76(8):1327-1339. doi:10.1136/annrheumdis-2016-209909; Маслянский АЛ, Мазуров ВИ, Зоткин ЕГ, Иливанова ЕП, Тотолян АА. Анти-В-клеточная терапия аутоиммунных заболеваний. Медицинская иммунология. 2007;9(1):15-34. doi:10.15789/1563-0625-2007-1-15-34; https://rsp.mediar-press.net/rsp/article/view/3188

  10. 10
    Academic Journal

    المصدر: Bulletin of Siberian Medicine; Том 21, № 3 (2022); 59-66 ; Бюллетень сибирской медицины; Том 21, № 3 (2022); 59-66 ; 1819-3684 ; 1682-0363 ; 10.20538/1682-0363-2022-21-3

    وصف الملف: application/pdf

    Relation: https://bulletin.tomsk.ru/jour/article/view/4907/3254; Smolen J.S., Aletaha D., Bijlsma J.W., Breedveld F.C., Boumpas D., Burmester G. et al. T2T Expert committee. Treating rheumatoid arthritis to target: recommendations of an international task force. Ann. Rheum. Dis. 2010;69(4):631–637. DOI:10.1136/ard.2009.123919.; Fransen J., Stucki G., van Riel P.L.C.M. Rheumatoid arthritis measures: Disease Activity Score (DAS), Disease Activity Score-28 (DAS28), Rapid Assessment of Disease Activity in Rheumatology (RADAR) and Rheumatoid Arthritis Disease Activity Index (RADAI). Arthritis & Rheumatism. 2003;49:214–224. DOI:10.1002/art.11407.; Prevoo M.L., van ‘t Hof M.A., Kuper H.H., van Leeuwen M.A., van de Putte L.B., van Riel P.L. Modified disease activity scores that include twenty-eight-joint counts. Development and validation in a prospective longitudinal study of patients with rheumatoid arthritis. Arthritis Rheum. 1995;38(1):44–48. DOI:10.1002/art.1780380107. PMID: 7818570.; De Jong P.H., Hazes J.M., van Zeben D., van der Lubbe P.A., de Jager M.H., de Sonnaville P.B. at al. Treatment decisions and related costs differ significantly depending on the choice of a disease activity index in RA, according to 1987 and 2010 classification criteria. Rheumatology (Oxford). 2012;51(7):1269– 1277. DOI:10.1093/rheumatology/kes008.; Marks J.L., Holroyd C.R., Dimitrov B.D., Armstrong R.D., Calogeras A., Cooper C. et al. Does combined clinical and ultrasound assessment allow selection of individuals with rheumatoid arthritis for sustained reduction of anti-tumor necrosis factor therapy? Arthritis Care Res. (Hoboken). 2015;67(6):746– 753. DOI:10.1002/acr.22552.; Miranda-García P., Chaparro M., Gisbert J.P. Correlation between serological biomarkers and endoscopic activity in patients with inflammatory bowel disease. Gastroenterol. Hepatol. 2016;39(8):508–515. DOI:10.1016/j.gastrohep.2016.01.015.; Korndörfer I.P., Brueckner F., Skerra A. The crystal structure of the human (S100A8/S100A9)2 heterotetramer, calprotectin, illustrates how conformational changes of interacting alpha-helices can determine specific association of two EF-hand proteins. J. Mol. Biol. 2007;370(5):887–898. DOI:10.1016/j.jmb.2007.04.065.; Romand X., Bernardy C., Nguyen M.V.C., Courtier A., Trocme C., Clapasson M. et al. Systemic calprotectin and chronic inflammatory rheumatic diseases. Joint Bone Spine. 2019;86(6):691–698. DOI:10.1016/j.jbspin.2019.01.003.; Lee D.G., Woo J.W., Kwok S.K., Cho M.L., Park S.H. MRP8 promotes Th17 differentiation via upregulation of IL-6 production by fibroblast-like synoviocytes in rheumatoid arthritis. Exp. Mol. Med. 2013;45(4):20. DOI:10.1038/ emm.2013.39.; Abildtrup M., Kingsley G.H., Scott D.L. Calprotectin as a biomarker for rheumatoid arthritis: a systematic review. J. Rheumatol. 2015;42(5):760–770. DOI:10.3899/jrheum.140628.; Bae S.C., Lee Y.H. Calprotectin levels in rheumatoid arthritis and their correlation with disease activity: a meta-analysis. Postgrad. Med. 2017;129(5):531–537. DOI:10.1080/00325481.2017.131972.; Hurnakova J., Zavada J., Hanova P., Hulejova H., Klein M., Mann H. et al. Serum calprotectin (S100A8/9): an independent predictor of ultrasound synovitis in patients with rheumatoid arthritis. Arthritis Res. Ther. 2015;17(1):252. DOI:10.1186/ s13075-015-0764-5.; Jarlborg M., Courvoisier D.S., Lamacchia C., Martinez Prat L., Mahler M. Bentow C. et al. Physicians of the Swiss Clinical Quality Management (SCQM) registry. Serum calprotectin: a promising biomarker in rheumatoid arthritis and axial spondyloarthritis. Arthritis Res. Ther. 2020;22(1):105. DOI:10.1186/s13075-020-02190-3.; Hammer H.B., Ødegård S., Syversen S.W., Lande wé R., van der Heijde D., Uhlig T. et al. Calprotectin (a major S100 leucocyte protein) predicts 10-year radiographic progression in patients with rheumatoid arthritis. Ann. Rheum. Dis. 2010;69(1):150–154. DOI:10.1136/ard.2008.103739.; Bach M., Moon J., Moore R., Pan T., Nelson J.L., Lood C. A neutrophil activation biomarker panel in prognosis and monitoring of patients with rheumatoid arthritis. Arthritis Rheumatol. 2020;72(1):47–56. DOI:10.1002/art.41062.; Altman R., Asch E., Bloch D., Bole G., Borenstein D., Brandt K. et al. Development of criteria for the classification and reporting of osteoarthritis. Classification of osteoarthritis of the knee. Diagnostic and Therapeutic Criteria Committee of the American Rheumatism Association. Arthritis Rheum. 1986;29(8):1039–1049. DOI:10.1002/art.1780290816.; https://bulletin.tomsk.ru/jour/article/view/4907

  11. 11
    Academic Journal

    المصدر: Rheumatology Science and Practice; Vol 59, No 4 (2021); 434–441 ; Научно-практическая ревматология; Vol 59, No 4 (2021); 434–441 ; 1995-4492 ; 1995-4484

    وصف الملف: application/pdf

    Relation: https://rsp.mediar-press.net/rsp/article/view/3061/2113; Насонов ЕЛ (ред.). Ревматология. Российские клинические рекомендации. М.:ГЭОТАР-Медиа;2017:464.; Qin B, Wang J, Yang Z, Yang M, Ma N, Huang F, et al. Epidemiology of primary Sjögren’s syndrome: A systematic review and meta-analysis. Ann Rheum Dis. 2015;74:1983-1989. doi:10.1136/annrheumdis-2014-205375; Shiboski CH, Shiboski SC, Seror R, Criswell LA, Labetoulle M, Lietman TM, et al. 2016 American College of Rheumatology/ European League Against Rheumatism classification criteria for primary Sjögren’s syndrome: A consensus and data-driven methodology involving three international patient cohorts. Arthritis Rheumatol. 2017;69:35-45. doi:10.1002/art.39859; Guellec D, Cornec D, Jousse-Joulin S, Marhadour T, Marcorelles P, Pers JO, et al. Diagnostic value of labial minor salivary gland biopsy for Sjögren’s syndrome: A systematic review. Autoimmun Rev. 2013;12:416-420. doi:10.1016/j.autrev.2012.08.001; Bodeutsch C, de Wilde PC, Kater L, van Houwelingen JC, van den Hoogen FH, Kruize AA, et al. Quantitative immunohistologic criteria are superior to the lymphocytic focus score criterion for the diagnosis of Sjögren’s syndrome. Arthritis Rheum. 1992;35:1075-1087. doi:10.1002/art.1780350913; van Woerkom JM, Kruize AA, Barendregt PJ, Kater L, Hené R, Bootsma H, et al. Clinical significance of quantitative immunohistology in labial salivary glands for diagnosing Sjogren’s syndrome. Rheumatology (Oxford). 2006;45:470-477. doi:10.1093/rheumatology/kei191; Fisher BA, Jonsson R, Daniels T, Bombardieri M, Brown RM, Morgan P, et al. Standardisation of labial salivary gland histopathology in clinical trials in primary Sjogren’s syndrome. Ann Rheum Dis. 2017;76:1161-1168. doi:10.1136/annrheumdis-2016-210448; Navazesh M, Kumar SKS. Measuring salivary flow. Dent Assist J. 2008;139:35S-40S. doi:10.14219/jada.archive.2008.0353; Teppo H, Revonta M. A follow-up study of minimally invasive lip biopsy in the diagnosis of Sjögren’s syndrome. Clin Rheumatol. 2007;26:1099-1103. doi:10.1007/s10067-006-0457-1; Quartuccio L, Baldini C, Bartoloni E, Priori R, Carubbi F, Corazza L, et al. Anti-SSA/SSB-negative Sjögren’s syndrome shows a lower prevalence of lymphoproliferative manifestations, and a lower risk of lymphoma evolution. Autoimmun Rev. 2015;14:1019-1022. doi:10.1016/j.autrev.2015.07.002; Christodoulou MI, Kapsogeorgou EK, Moutsopoulos HM. Characteristics of the minor salivary gland infiltrates in Sjögren’s syndrome. J Autoimmun. 2010;34:400–407. doi:10.1016/j.jaut.2009.10.004; Jonsson MV, Skarstein K. Follicular dendritic cells confirm lymphoid organization in the minor salivary glands of primary Sjögren’s syndrome. J Oral Pathol Med. 2008;37:515-521. doi:10.1111/j.1600-0714.2008.00674.x; Braun M, Melchers I, Peter HH, Illges H. Human B and T lymphocytes have similar amounts of CD21 mRNA, but differ in surface expression of the CD21 glycoprotein. Int Immunol. 1998;10(8):1197-1202.; Daridon C, Pers JO, Devauchelle V, Martins-Carvalho C, Hutin P, Pennec YL, et al. Identification of transitional type II B cells in the salivary glands of patients with Sjögren’s syndrome. Arthritis Rheum. 2006;54:2280-2288. doi:10.1002/art.21936; Le Pottier L, Devauchelle V, Fautrel A, Daridon C, Saraux A, Youinou P, et al. Ectopic germinal centers are rare in Sjogren’s syndrome salivary glands and do not exclude autoreactive B cells. J Immunol. 2009;182:3540-3547. doi:10.4049/jimmunol.0803588; Ushio A, Arakaki R, Yamada A, Saito M, Tsunematsu T, Kudo Y, et al. Crucial roles of macrophages in the pathogenesis of autoimmune disease. World J Immunol. 2017;7(1):1-8. doi:10.5411/wji.v7.i1.1; Ushio A, Arakaki R, Otsuka K, Yamada A, Tsunematsu T, Kudo Y, et al. CCL22-producing resident macrophages enhance T cell response in Sjögren’s syndrome. Front Immunol. 2018;9:2594. doi:10.3389/fimmu.2018.02594; https://rsp.mediar-press.net/rsp/article/view/3061

  12. 12
    Academic Journal

    المصدر: Rheumatology Science and Practice; Vol 58, No 6 (2020); 658-662 ; Научно-практическая ревматология; Vol 58, No 6 (2020); 658-662 ; 1995-4492 ; 1995-4484

    وصف الملف: application/pdf

    Relation: https://rsp.mediar-press.net/rsp/article/view/2963/2037; Эрдес Ш, Бадокин ВВ, Бочкова АГ, Бугрова ОВ, и др. О терминологии спондилоартритов. Научно-практическая ревматология. 2015;53(6):657-660. DOI:10.14412/1995-4484-2015-657-660; Эрдес Ш, Ребров АП, Дубинина ТВ, и др. Спондилоартриты: современная терминология и определения. Терапевтический архив. 2019;91(5):84-88. DOI:10.26442/00403660.2019.05.000208; Rudwaleit M, van der Heijde D, Landewc R, et al. The development of Assessment of SpondyloArthritis international Society classification criteria for axial spondyloarthritis (part II): validation and final selection. Ann Rheum Dis. 2009;68(6):777-783. DOI:10.1136/ard.2009.108233; Maksymowych WP. Biomarkers for diagnosis of axial spondyloar-thritis, disease activity, prognosis, and prediction of response to therapy. Front Immunol. 2019;10:305. DOI:10.3389/fimmu.2019.00305; Baraliakos X, Baerlecken N, Witte T, Heldmann F, Braun J. High prevalence of anti-CD74 antibodies specific for the HLA class II-associated invariant chain peptide (CLIP) in patients with axial spondyloarthritis. Ann Rheum Dis. 2014;73(6):1079-1082. DOI:10.1136/annrheumdis-2012-202177; Starlets D, Gore Y, Binsky I, et al. Cell-surface CD74 initiates a signaling cascade leading to cell proliferation and survival. Blood. 2006;107(12):4807-4816. DOI:10.1182/blood-2005-11-4334; Ranganathan V, Ciccia F, Zeng F, et al. Macrophage migration inhibitory factor induces inflammation and predicts spinal progression in ankylosing spondylitis. Arthritis Rheumatol. 2017;69(9):1796-1806. DOI:10.1002/art.40175; Liu Y, Liao X, Shi G. Autoantibodies in spondyloarthritis, focusing on anti-CD74 antibodies. Front Immunol. 2019;10:5. DOI:10.3389/fimmu.2019.00005; Garrett S, Jenkinson T, Kennedy LG, Whitelock H, Gaisford P, Calin A. A new approach to defining disease status in ankylosing spondylitis: the Bath Ankylosing Spondylitis Disease Activity Index. J Rheumatol. 1994;21(12):2286-2291.; Lukas C, I .andewc R, Sieper J, et al. Assessment of SpondyloArthritis international Society. Development of an ASAS-endorsed disease activity score (ASDAS) in patients with ankylosing spondylitis. Ann Rheum Dis. 2009;68(1):18-24. DOI:10.1136/ard.2008.094870; Кузнецова ДА, Лапин СВ, Гайдукова ИЗ, Ребров АП, Маслянский АЛ. Клинико-диагностическая значимость аутоантител к CD74 при аксиальных спондилоартритах. Клиническая лабораторная диагностика. 2018;63(5):297-301. DOI:10.18821/0869-2084-2018-63-5-297-301; Baerlecken NT, Nothdorft S, Stummvoll GH, et al. Autoantibodies against CD74 in spondyloarthritis. Ann Rheum Dis. 2014;73(6):1211-1214. DOI:10.1136/annrheumdis-2012-202208; Riechers E, Baerlecken N, Baraliakos X, et al. Sensitivity and specificity of autoantibodies against CD74 in nonradiographic axial spondyloarthritis. Arthritis Rheumatol. 2019;71(5):729-735. DOI:10.1002/art.40777; Abdelaziz MM, Gamal RM, Ismail NM, Lafy RA, Hetta HF. Diagnostic value of anti-CD74 antibodies in early and late axial spondyloarthritis and its relationship to disease activity. Rheumatology. Published online July 24, 2020. DOI:10.1093/rheumatology/keaa292; De Craemer AS, Witte T, Deroo L, et al. FRI0312 anti-CD74 IgA antibodies are most sensitive and specific to identify young male axial spondyloarthritis patients. Ann Rheum Dis. 2020;79:746-747.; Ziade NR, Mallak I, Merheb G, Ghorra P, Baerlecken N, Witte T, et al. Added Value of anti-CD74 autoantibodies in axial spondy-loarthritis in a population with low HLA-B27 prevalence. Front Immunol. 2019;10:574. DOI:10.3389/fimmu.2019.00574; de Winter JJ, van de Sande MG, Baerlecken N, et al. Anti-CD74 antibodies have no diagnostic value in early axial spondyloarthritis: data from the spondyloarthritis caught early (SPACE) cohort. Arthritis Res Ther. 2018;20(1):38. DOI:10.1186/s13075-018-1535-x; https://rsp.mediar-press.net/rsp/article/view/2963

  13. 13
    Academic Journal

    المصدر: Rheumatology Science and Practice; Vol 55, No 2 (2017); 159-163 ; Научно-практическая ревматология; Vol 55, No 2 (2017); 159-163 ; 1995-4492 ; 1995-4484 ; 10.14412/rsp20172

    وصف الملف: application/pdf

    Relation: https://rsp.mediar-press.net/rsp/article/view/2357/1560; Krecic AM, Swanson MS. HnRNP complexes: composition, structure, and function. Curr Opin Cell Biol. 1999;11(3):363-71. doi:10.1016/S0955-0674(99)80051-9; Swanson MS. Functions of nuclear pre-mRNA/mRNA binding proteins. In: Lamond AI, editor. Pre-mRNA Processing. Berlin, Heidelberg: Landes Co Springer Verlag; 1995. P. 17-33.; Maslyanskiy AL, Olinek PA, Lapin SV, et al. Anti-hnRNP B1 (RA33) autoantibodies are associated with the clinical phenotype in russian patients with rheumatoid arthritis and systemic sclerosis. J Immunol. 2014;2014:Article ID 51659. doi:10.1155/2014/516593; Олейник ПА, Маслянский АЛ, Лапин СВ и др. Антитела к HnRNP (RA33) у больных с ревматоидный артритом. Медицинский академический журнал. 2014;14(3):59-66 [Oleinik PA, Maslyanskiy AL, Lapin SV, et al. Anti-HNRNP (RA33) antibody in rheumatoid arthritis. Meditsinskii Akademicheskii Zhurnal. 2014;14(3):59-66 (In Russ.)].; Aletaha D, Neogi T, Silman AJ, et al. 2010 Rheumatoid arthritis classification criteria: an American College of Rheumatology. European League Against Rheumatism collaborative initiative. Ann Rheum Dis. 2011;69:1580-8. doi:10.1136/ard.2010.138461; Meyer O, Tauxe F, Fabregas D, et al. Anti-RA33 antinuclear antibody in rheumatoid arthritis and mixed connective tissue disease: comparison with antikeratin and antiperinuclear antibodies. Clin Exp Rheumatol. 1993;11:473-8.; Лапин СВ, Маслянский АЛ. Лабораторная диагностика ревматоидного артрита. Новые перспективы. Клинико-лабораторный консилиум. 2009;1(26):69-74 [Lapin SV, Maslyansky AL. Laboratory diagnosis of rheumatoid arthritis: new perspectives. Kliniko-Laboratornyi Konsilium. 2009;1(26):69-74 (In Russ.)].; Van der Linden S, Valkenburg HA, Cats A. Evaluation of diagnostic criteria for ankylosing spondylitis. A proposal for modification of the New York criteria. Arthritis Rheum. 1984 Apr;27(4):361. doi:10.1002/art.1780270401; American College of Rheumatology Guidelines for Screening, Treatment, and Management of Lupus Nephritis. Arthritis Care Res. 2012 Jun;64(6):797-808. doi:10.1002/acr.21664; Masi AT, Rodnan GP, Medsger TA, et al. Preliminary criteria for the classification of systemic sclerosis (scleroderma). Arthritis Rheum. 1980;23:581-90. doi:10.1002/art.1780230510 11. Васильев ВИ, Симонова МВ, Сафонова ТН. Критерии диагноза болезни и синдрома Шегрена. В кн.: Насонова ВА, Бунчук НВ, редакторы. Избранные лекции по клинической ревматологии. Москва: Медицина; 2001. С. 112-132 [Vasil'ev VI, Simonova MV, Safonova TN. Criteria for the diagnosis of Sjogren's disease and syndrome. In: Nasonova VA, Bunchuk NV, editors. Izbrannye lektsii po klinicheskoi revmatologii (Selected lectures on clinical rheumatology). Moscow: Meditsina; 2001. P. 112-132]. 12. Лапин СВ, Тотолян АА. Иммунологическая лабораторная диагностика аутоимунных заболеваний. Санкт-Петербург: Человек; 2010. 272 с. [Lapin SV, Totolyan AA. Immunologicheskaya laboratornaya diagnostika autoimunnykh zabolevanii (Immunological laboratory diagnostics of autoimmune diseases). Sankt-Peterburg: Chelovek; 2010. 272 p.]. 13. Sung Bin Cho, Keun Jae Ahn, Do Hee Kim, et al. Identification of HnRNP-A2/B1 as a Target Antigen of Anti-Endothelial Cell IgA Antibody in Behchet's Disease. J Invest Dermatol. 2012;132:601-8. doi:10.1038/jid.2011.397; https://rsp.mediar-press.net/rsp/article/view/2357

  14. 14
    Academic Journal

    المصدر: Medical Immunology (Russia); Том 19, № 2 (2017); 175-184 ; Медицинская иммунология; Том 19, № 2 (2017); 175-184 ; 2313-741X ; 1563-0625 ; 10.15789/1563-0625-2017-2

    وصف الملف: application/pdf

    Relation: https://www.mimmun.ru/mimmun/article/view/1220/912; Байдун Л.А., Зурочка А.В., Тотолян Арег А., Хайдуков С.В. Стандартизованная технология «Исследование субпопуляционного состава лимфоцитов периферической крови с применением Проточных цитофлюориметров-анализаторов» (проект)// Медицинская иммунология, 2012. Т. 14, № 3. С.255-268. [Baydun L.A., Zurochka A.V., Totolian Areg A., Khaydukov S.V. Standardized technology “Peripheral blood Lymphocyte subpopulations by using of flow cytometry-analizators” (proect). Meditsinskaya immunologiya = Medical Immunology (Russia), 2012, Vol. 14, no. 3, pp. 255-268. [In Russ.] http://dx.doi. org/10.15789/1563-0625-2012-3-255-268; Кудрявцев И.В., Субботовская А.И. Опыт измерения параметров иммунного статуса с использованием шестицветного цитофлуоримерического анализа // Медицинская иммунология, 2015. T. 17, № 1. С. 19-26. [Kudryavtsev I.V., Subbotovskaya A.I. Application of six-color flow cytometric analysis for immune profile monitoring. Meditsinskaya immunologiya = Medical Immunology (Russia), 2015, Vol. 17, no. 1, pp.19-26. [In Russ.] http://dx.doi.org/10.15789/1563-0625- 2015-1-19-26; Лазарева Н.М., Лапин С.В., Мазинг А.В., Булгакова Т.В., Иливанова Е.П., Маслянский А.Л., Тотолян А.А. Оптимизация комплекса серологических методов диагностики системных заболеваний соединительной ткани // Клиническая лабораторная диагностика, 2011. № 12. С. 12-17. [Lazareva N.M., Lapin S.V., Mazing A.V., Bulgakova T.V., Ilivanova E.P., Maslyansky A.L., Totolian A.A. Optimization of serological diagnostic methods of connective tissue diseases. Klinicheskaya laboratornaya diagnostika = Clinical Laboratory Diagnostics, 2011, no. 12, pp. 12-17. [In Russ.]; Маслянский А.Л., Пенин И.Н., Чешуина М.Д., Тришина И.Н., Новикова А.Н., Колесова Е.П., Лазарева Н.М., Мазинг А.В., Лапин С.В., Малышкин К.А., Сысоев К.А., Мазуров В.И., Конради А.О., Назаров П.Г. Общие закономерности продукции цитокинов и хемокинов у больных диффузными заболеваниями соединительной ткани, воспалительными артропатиями и атеросклерозом // Цитокины и воспаление, 2014. Т. 13, № 3. С. 9-21. [Maslyanskiy A.L., Penin I.N., Cheshuina M.D., Trichina I.N., Novikova A.N.,Kolesova E.P., Lazareva N.M., Mazing A.V., Lapin S.V., Malishkin K.A., Sysoev К.А., Mazurov V.I., Konradi А.О., Nazarov P.G. Common consistent patterns of the cytokine and chemokine production in patients with diffuse connective tissue diseases, inflammatory arthropathies and atherosclerosis. Tsitokiny i vospalenie = Cytokines and Inflammation, 2014, Vol. 13, no. 3, pp. 9-21. [In Russ.]; Насонов E. Л.,Соловьев С. К. Перспективы применения моноклональных антител к В-лимфоцитам (ритуксимаб) при воспалительных ревматических заболеваниях // Научно-практическая ревматология, 2007. № 1. С. 4-8. [Nasonov E.L., Solovyev S.K. Perspectives of monoclonal anti-B lymphocyte antibodies (rituximab) administration in inflammatory rheumatic diseases. Nauchno-prakticheskaya revmatologiya = Rheumatology Science and Practice, 2007, no. 1, pp. 4-8. [In Russ.]; Созина А.В., Неустроева Ю.А., Тихомирова Т.А., Лапин С.В. Сочетанная встречаемость аутоантител у больных с диффузными болезнями соединительной ткани // Медицинская иммунология, 2007. Т. 9, № 1. С. 61- 68. [Sozina A.V., Neustroeva U.A., Tihomirova T.A., Lapin S.V. Coincidence of autoantibodies among patients with diffuse connective tissue disorders. Meditsinskaya immunologiya = Medical Immunology (Russia), 2007, Vol. 9, no. 1, pp. 61-68. [In Russ.] http://dx.doi.org/10.15789/1563-0625-2007-1-61-68; Ярилин А.А. Иммунология. М.: ГЭОТАР-Медиа, 2010. C. 354-479. [Yarilin AA. Immunology]. Moscow: GEOTARMedia, 2010, pp. 354-479.; Arce E., Jackson D.G., Gill M.A, Bennett L.B., Banchereau J., Pascual V. Increased frequency of pre-germinal center B cells and plasma cell precursors in the blood of children with systemic lupus erythematosus. The Journal of Immunology, 2001, Vol. 8, p. 167.; Binard A., Le Pottier L., Devauchelle V., Saraux A., Youinou P., Pers J.O. Is the blood B-cell subset profile diagnostic for SjÖgren syndrome? Annals of the Rheumatic Diseases, 2009, Vol. 68, p. 447.; Bohnhorst J.Q., Bjørgan M.B., Thoen J.E, Natvig J.B., Thompson K.M. Bm1-Bm5 classification of peripheral blood B cells reveals circulating germinal center founder cells in healthy individuals and disturbance in the B cell subpopulations in patients with primary Sjögren’s syndrome. The Journal of Immunology, 2001, Vol. 10, p. 167.; Cancro M.P., Hao Y., Scholz J.L., Riley R.L., Frasca D., Dunn-Walters D.K., Blomberg B.B. B cells and aging: molecules and mechanisms. Trends Immunol., 2009, Vol. 30, no. 7, pp. 313-318.; Dörner T., Jacobi A.M., Lipsky P.E. B cells in autoimmunity. Arthritis Research & Therapy, 2009, Vol. 11, no. 5, p. 247.; Duchamp M. B-cell subpopulations in children: national reference values. Immunity, Inflammation and Disease, 2014, Vol. 11, pp. 131-140.; Fassbinder T., Saunders U., Mickholz E., Jung E., Becker H., Schlüter B., Jacobi A.M. Differential effects of cyclophosphamide and mycophenolate mofetil on cellular and serological parameters in patients with systemic lupus erythematosus. Arthritis Research & Therapy, 2015, Vol. 17, no. 1, p. 92.; Glaesener S., Quách T.D., Onken N., Weller-Heinemann F., Dressler F., Huppertz H.I., Thon A., Meyer-Bahlburg A. Distinct effects of methotrexate and etanercept on the b cell compartment in patients with juvenile idiopathic arthritis. Arthritis & Rheumatology, 2014, Vol. 66, no. 9, pp. 2590-2600.; Hampe C.S. B cells in autoimmune diseases. Scientifica, 2012, Vol. 9, pp. 1-18.; Iwata S., Saito K., Tokunaga M., Yamaoka K., Nawata M., Yukawa S., Hanami K., Fukuyo S., Miyagawa I., Kubo S., Tanaka Y. Phenotypic changes of lymphocytes in patients with systemic lupus erythematosus who are in longterm remission after B cell depletion therapy with rituximab. The Journal of Rheumatology, 2011, Vol. 38, no. 4, pp. 633-641.; Jacobi A.M., Odendahl M., Reiter K., Bruns A., Burmester G.R., Radbruch A., Valet G,. Lipsky P.E., Dorner T. Correlation between circulating cd27high plasma cells and disease activity in patients with systemic lupus erythematosus. Arthritis & Rheumatism, 2003, Vol. 48, no. 5, pp. 1332-1342.; Jacobi A.M., Weiqing W.H., Tao, Freimuth W., Inaki S., Furie R., Mackay M., Aranow C., Diamond B., Davidson A. The effect of prolonged treatment with belimumab on b cells in human SLE. Arthritis & Rheumatology, 2010, Vol. 62, no. 1, pp. 201- 210.; Leandro M.J. B-cell subpopulations in humans and their differential susceptibility to depletion with anti-CD20 monoclonal antibodies. Arthritis Research & Therapy, 2013, Vol. 15, Suppl 1, S3.; Mahnke Y.D., Roederer M. Optimizing a multicolor immunophenotyping assay. Clinics in Laboratory Medicine, 2007, Vol. 27, pp. 469-485.; Mariele Gatto, Emese Kiss, Yaakov Naparstek, and Andrea Doria. In-/off-label use of biologic therapy in systemic lupus erythematosu. BMC Medicine, 2014, Vol. 12, p. 30.; Odendahl M., Keitzer R, Wahn U., Hiepe F., Radbruch A., Dörner T., Bunikowski R. Perturbations of peripheral b lymphocyte homoeostasis in children with systemic lupus erythematosus. Annals of the Rheumatic Diseases, 2003, Vol. 62, pp. 851-858.; Pers J.O., Youinou P. Are the B cells cast with the leading part in the Sjögren’s syndrome scenario? Oral Diseases, 2014, Vol. 20, no. 6, pp. 529-537.; Petri M., Orbai A.M., Alarcon G.S., Gordon C., Merrill J.T., Fortin P.R., et al. Derivation and validation of the Systemic Lupus International Collaborating Clinics classification criteria for systemic lupus erythematosus. Arthritis & Rheumatology , 2012, Vol. 64, no. 8, pp. 2677-2686.; Prak Luning E.T., Ross J., Sutter J., Sullivan K.E. Age-related trends in pediatric B-cell subsets. Pediatric and Developmental Pathology, 2011, Vol. 14, no. 1, pp. 45-52.; Sanz I., Wei C., Lee F.E., Anolik J. Phenotypic and functional heterogeneity of human memory B cells. Seminars in Immunology, 2008, Vol. 20, no. 1, pp. 67-82.; Vale A.M., Kearney J.F., Nobrega Alberto, Schroeder H.W. Molecular biology of B cells. 3rd ed. 2015, pp. 99-119.; Vences-Catalan F., Santos-Argumedo L. CD38 Through the life of a murine B lymphocyte. IUBMB Life, 2011, Vol. 63, no. 10, pp. 840-846.; Yanaba K., Bouaziz J.D., Matsushita T., Magro C.M., St. Clair E.W, Tedder T.F. B-lymphocyte contributions to human autoimmune disease. Immunological Reviews, 2008, Vol. 223, no. 6, pp.284-299; https://www.mimmun.ru/mimmun/article/view/1220

  15. 15
    Academic Journal

    المصدر: Rheumatology Science and Practice; Vol 54, No 1 (2016); 10-15 ; Научно-практическая ревматология; Vol 54, No 1 (2016); 10-15 ; 1995-4492 ; 1995-4484 ; 10.14412/rsp20161

    وصف الملف: application/pdf

    Relation: https://rsp.mediar-press.net/rsp/article/view/2163/1385; Каратеев ДЕ, Александрова ЕН, Демидова ИВ и др. Антицитруллиновые антитела и данные магнитно-резонансной томографии суставов кисти у больных ранним артритом. Терапевтический архив. 2008;80(10):72-6 [Karateev DE, Aleksandrova EN, Demidova IV, et al. Anticitrullin antibodies and data of magnetic resonance imaging of hand joints in patients with early arthritis. Terapevticheskii arkhiv. 2008;80(10):72-6 (In Russ.)].; Жебрун ДА, Тотолян АА, Маслянский АЛ и др. Синтез ангиогенных и ангиостатических СXС-хемокинов и их рецепторов в синовиальной оболочке при ревматоидном артрите. Цитокины и воспаление. 2014;13(2):39-44 [Zhebrun DA, Totolyan AA, Maslyanskii AL, et al. Synthesis of endogenous angiostatic and CXC chemokines and their receptors in synovium in rheumatoid arthritis. Tsitokiny i vospalenie. 2014;13(2):39-44 (In Russ.)].; Altman R, Asch E, Bloch D, et al. Development of criteria for the classification and reporting of osteoarthritis. Classification of osteoarthritis of the knee. Diagnostic and Therapeutic Criteria Committee of the American Rheumatism Association. Arthritis Rheum. 1986;29(8):1039-49. doi:10.1002/art.1780290816; Жебрун ДА, Маслянский АЛ, Титов АГ и др. Определение мРНК ангиогенных и ангиостатических хемокинов и их рецепторов в синовиальной оболочке методом количественной ПЦР в реальном времени. Медицинская иммунология. 2013;15(6):525-34 [Zhebrun DA, Maslyanskii AL, Titov AG, et al. Determination of mRNA of angiogenic and angiostatic chemokines and their receptors in the synovium by quantitative real-time PCR. Meditsinskaya immunologiya. 2013;15(6):525-34 (In Russ.)]. doi: /10.15789/1563-0625-2013-6-525-534; Corsiero E, Bombardieri M, Manzo A, et al. Role of lymphoid chemokines in the development of functional ectopic lymphoid structures in rheumatic autoimmune diseases. Immunol Lett. 2012;145(1-2):62-7. doi:10.1016/j.imlet.2012.04.013; Kokebie R, Aggarwal R, Lidder S, et al. The role of synovial fluid markers of catabolism and anabolism in osteoarthritis, rheumatoid arthritis and asymptomatic organ donors. Arthritis Res Ther. 2011;13(2):R50. doi:10.1186/ar3293; Маслянский АЛ, Лапин СВ, Мазинг АВ и др. Диагностическая значимость серологических маркеров ревматоидного артрита. Научно-практическая ревматология. 2012;50(5):20-4 [Maslaynski AL, Lapin SV, Mazing AV, et al. Diagnostic value of serological markers of rheumatoid arthritis. Nauchno-prakticheskaya revmatologiya = Rheumatology Science and Practice. 2012;50(5):20-4 (In Russ.)]. doi:10.14412/1995-4484-2012-1175; Маслянский АЛ, Мазуров ВИ, Зоткин ЕГ и др. Анти-В-клеточная терапия аутоиммунных заболеваний. Медицинская иммунология. 2007;9(1):15-34 [Masliansky AL, Mazurov VI, Zotkin EG, et al. Anti-B-cell therapy of autoimmune diseases. Meditsinskaya immunologiya = Medical Immunology. 2007;9(1):15-34 (In Russ.)]. doi:10.15789/1563-0625-2007-1-15-34; Humby F, Bombardieri M, Manzo A, et al. Ectopic lymphoid structures support ongoing production of class-switched autoantibodies in rheumatoid synovium. PLoS Med. 2009;6(1):e1. doi:10.1371/journal.pmed.0060001; Bugatti S, Manzo A, Vitolo B, et al. High expression levels of the B cell chemoattractant CXCL13 in rheumatoid synovium are a marker of severe disease. Rheumatology (Oxford). 2014;53(10):1886-95. doi:10.1093/rheumatology/keu163; Grassi F, Cristino S, Toneguzzi S, et al. CXCL12 chemokine upregulates bone resorption and MMP-9 release by human osteoclasts: CXCL12 levels are increased in synovial and bone tissue of rheumatoid arthritis patients. J Cell Physiol. 2004;199(2):244-51. doi:10.1002/jcp.10445; Hansen IB, Ellingsen T, Hornung N, et al. Plasma level of CXC-chemokine CXCL12 is increased in rheumatoid arthritis and is independent of disease activity and methotrexate treatment. J Rheumatol. 2006;33(9):1754-9.; Loeuille D, Chary-Valckenaere I, Champigneulle J, et al. Macroscopic and microscopic features of synovial membrane inflammation in the osteoarthritic knee: correlating magnetic resonance imaging findings with disease severity. Arthritis Rheum. 2005;52(11):3492-501. doi:10.1002/art.21373; Chen B, Deng Y, Tan Y, et al. Association between severity of knee osteoarthritis and serum and synovial fluid interleukin 17 concentrations. J Int Med Res. 2014;42(1):138-44. doi:10.1177/0300060513501751; Hampel U, Sesselmann S, Iserovich P, et al. Chemokine and cytokine levels in osteoarthritis and rheumatoid arthritis synovial fluid. J Immunol Methods. 2013;396(1-2):134-9. doi:10.1016/j.jim.2013.08.007; Borzi RM, Mazzetti I, Macor S, et al. Flow cytometric analysis of intracellular chemokines in chondrocytes in vivo: constitutive expression and enhancement in osteoarthritis and rheumatoid arthritis. FEBS Lett. 1999;455(3):238-42. doi:10.1016/S0014-5793(99)00886-8; Kanbe K, Takagishi K, Chen Q. Stimulation of matrix metalloprotease 3 release from human chondrocytes by the interaction of stromal cell-derived factor 1 and CXC chemokine receptor 4. Arthritis Rheum. 2002;46(1):130-7. doi:10.1002/1529-0131(200201)46:13.0.CO;2-D; Lisignoli G, Cristino S, Toneguzzi S, et al. IL1beta and TNFalpha differently modulate CXCL13 chemokine in stromal cells and osteoblasts isolated from osteoarthritis patients: evidence of changes associated to cell maturation. Exp Gerontol. 2004;39(4):659-65. doi:10.1016/j.exger.2003.09.030; Lee SS, Joo YS, Kim WU, et al. Vascular endothelial growth factor levels in the serum and synovial fluid of patients with rheumatoid arthritis. Clin Exp Rheumatol. 2001;19(3):321-4.; Haywood L, McWilliams DF, Pearson CI, et al. Inflammation and angiogenesis in osteoarthritis. Arthritis Rheum. 2003;48(8):2173-7. doi:10.1002/art.11094; https://rsp.mediar-press.net/rsp/article/view/2163

  16. 16
    Academic Journal

    المصدر: Modern Rheumatology Journal; Том 10, № 1 (2016); 31-36 ; Современная ревматология; Том 10, № 1 (2016); 31-36 ; 2310-158X ; 1996-7012 ; 10.14412/1996-7012-2016-1

    وصف الملف: application/pdf

    Relation: https://mrj.ima-press.net/mrj/article/view/668/654; Massimo I, Yehuda A. Management of pericardial effusion. Eur Heart J. 2013 Apr;34(16):1186-97. doi:10.1093/eurheartj/ehs372. Epub 2012 Nov 2.; Maisch B, Seferovic PM, Risti AD, et al. Task Force on the Diagnosis and Management of Pericardial Diseases of the European Society of Cardiology. Guidelines on the diagnosis and management of pericardial diseases executive summary: the Task force on the diagnosis and management of pericardial diseases of the European Society of Cardiology. Eur Heart J. 2004 Apr;25(7):587-610.; Maestroni S, Di Corato PR, Cumetti D, et al. Recurrent pericarditis: autoimmune or autoinflammatory? Autoimmun Rev. 2012 Nov;12(1):60-5. doi:10.1016/j.autrev.2012. 07.023. Epub 2012 Aug 2.; Салугина СО, Федоров ЕС, Кузьмина НН, Захарова ЕЮ. Ингибитор интерлейкина 1 канакинумаб в лечении криопирин-ассоциированных периодических синдромов (CAPS). Клинический опыт. Современная ревматология. 2014;8(4):17-24. [Salugina SO, Fedorov ES, Kuz'mina NN, Zakharova EYu. The interleukin 1 inhibitor canakinumab in the treatment of cryopyrin-associated periodic syndromes (CAPS): clinical experience. Sovremennaya revmatologiya = Modern Rheumatology Journal. 2014;8(4):17-24. (In Russ.)]. DOI: http://dx.doi.org/10.14412/1996-7012-2014-4-17-24; Федоров ЕС, Салугина СО, Кузьмина НН. Аутовоспалительные синдромы: что необходимо знать ревматологу. Современная ревматология. 2012;6(2): 49-59. [Fedorov ES, Salugina SO, Kuz'mina NN. Autoinflammatory syndromes: What a rheumatologist should know. Sovremennaya revmatologiya = Modern Rheumatology Journal. 2012;6(2):49-59. (In Russ.)]. DOI: http://dx.doi.org/10.14412/1996-7012-2012-728; Fugii T, Akzuki M, Kameda H, et al. Methotrexate treatment in patients with adult onset Still’s disease: retrospective study of 13 Japanese cases. Ann Rheum Dis. 1997 Feb;56(2):144-8.; Sakai R, Nagasawa H, Nishi E, et al. Successful treatment of adult-onset Still's disease with tocilizumab monotherapy: two case reports and literature review. Clin Rheumatol. 2012 Mar;31(3):569-74. doi:10.1007/s10067-011-1917-9. Epub 2012 Jan 4.; Mahroum N, Mahagna H, Amital H. Diagnosis and classification of adult Still’s disease. J Autoimmun. 2014 Feb-Mar; 48-49:34-7. doi:10.1016/j.jaut.2014.01.011. Epub 2014 Jan 31.; Yamaguchi M, Ohta A, Tsunematsu T, et al. Preliminary criteria for classification of adult Still’s disease. J Rheumatol. 1992 Mar;19(3):424-30.; Fautrel B. Adult-onset Still disease. Best Pract Res Clin Rheumatol. 2008 Oct;22(5):773-92. doi:10.1016/j.berh.2008.08.006.; Hochberg MC. MACP Rheumatology. Sixth Edition. Mosby Elsevier; 2015. Chapter 87. P. 720-6.; Ichida H, Kawaguchi Y, Sugiura T, et al. Clinical manifestations of Adult-onset Still's disease presenting with erosive arthritis: Association with low levels of ferritin and Interleukin-18. Arthritis Care Res (Hoboken). 2014 Apr;66(4):642-6.; Garcia-Garcia G1, Fernandez-Auzmendi V, Olgado-Ferrero F, et al. Acute Miopericarditis as the Presenting Feature of Adult-Onset Still’s Disease. Reumatol Clin. 2012 Jan-Feb;8(1):31-3. doi:10.1016/j.reuma.2011.03.002. Epub 2011 Jun 17.; Yoo WH. Adult onset Still's disease flared with pericardial effusion. Rheumatol Int. 2008 Jan;28(3):285-7. Epub 2007 Aug 1.; Maria AT, Le Quellec A, Jorgensen C, et al. Adult onset Still's disease (AOSD) in the era of biologic therapies: Dichotomous view for cytokine and clinical expressions. Autoimmun Rev. 2014 Nov;13(11):1149-59. doi:10.1016/j.autrev.2014.08.032. Epub 2014 Aug 27.; https://mrj.ima-press.net/mrj/article/view/668

  17. 17
    Academic Journal

    المساهمون: грант Минобрнауки России и грант Правительства Санкт-Петербурга

    المصدر: Medical Immunology (Russia); Том 15, № 6 (2013); 525-534 ; Медицинская иммунология; Том 15, № 6 (2013); 525-534 ; 2313-741X ; 1563-0625 ; 10.15789/1563-0625-2013-6

    وصف الملف: application/pdf

    Relation: https://www.mimmun.ru/mimmun/article/view/676/724; Arenberg D.A., Keane M.P., DiGiovine B., Kunkel S.L., Morris S.B., Xue Y.Y., Burdick M.D., Glass M.C., Iannettoni M.D., Strieter R.M. Epithelial neutrophil activating peptide (ENA-78) is an important angiogenic factor in non-small cell lung cancer. J. Clin Invest., 1998, vol. 102, pp. 465-472.; Blades M.C., Ingegnoli F., Wheller S.K., Manzo A., Wahid S., Panayi G.S., Perretti M., Pitzalis C. Stromal cell-derived factor 1 (CXCL12) induces monocyte migration into human synovium transplanted onto SCID Mice. Arthritis Rheum., 2002, vol. 46, no. 3, pp. 824-836.; Boulday G., Haskova Z., Reinders M.E., Pal S., Briscoe D.M. Vascular endothelial growth factorinduced signaling pathways in endothelial cells that mediate overexpression of the chemokine IFN-gammainducible protein of 10 kDa in vitro and in vivo. J. Immunol., 2006, vol. 176, pp. 3098-3107.; Bustin S.A., Nolan T. Pitfalls of quantitative real-time reverse-transcription polymerase chain reaction. J. Biomol. Tech., 2004, vol. 15, pp. 155-166.; Chen Z., Malhotra P.S., Thomas G.R., Ondrey F.G., Duffey D.C., Smith C.W., Enamorado I., Yeh N.T., Kroog G.S., Rudy S., McCullagh L., Mousa S., Quezado M., Herscher L.L., Van Waes C. Expression of proinflammatory and proangiogenic cytokines in patients with head and neck cancer. Clin. Cancer Res., 1999, vol. 5, pp. 1369-1379.; Esche C., Stellato C., Beck L.A. Chemokines: key players in innate and adaptive immunity. J. Invest. Dermatol., 2005, vol. 125, pp. 615-628.; Firestein G.S. Etiology and pathogenesis of rheumatoid arthritis. Kelly’s Textbook of Rheumatology. Philadelphia, PA, Saunders Elsevier, 2009, pp. 1035-1086.; Frangogiannis N.G., Entman M.L. Chemokines in myocardial ischemia. Trends Cardiovasc. Med., 2005, vol. 15, pp. 163-169.; Heid C.A., Stevens J., Livak K.J., Williams P.M. Real time quantitative PCR. Genome Res.,1996, vol. 6, no. 10, pp. 986-994.; Hellemans J., Mortier G., De Paepe A., Speleman F., Vandesompele J. qBase relative quantification framework and software for management and automated analysis of real-time quantitative PCR data. Genome Biology, 2007, vol. 8, p. R19.; Hitchon C.A., Alex P., Erdile L.B., Frank M.B., Dozmorov I., Tang Y., Wong K., Centola M., El- Gabalawy H.S. A distinct multicytokine profile is associated with anti-cyclical citrullinated peptide antibodies in patients with early untreated inflammatory arthritis. J. Rheumatol., 2004 Dec, vol. 31, no. 12, pp. 2336-2346.; Keane M.P., Donnelly S.C., Belperio J.A., Goodman R.B., Dy M., Burdick M.D., Fishbein M.C., Strieter R.M. Imbalance in the expression of CXC chemokines correlates with bronchoalveolar lavage fluid angiogenic activity and procollagen levels in acute respiratory distress syndrome. J. Immunol., 2002, vol. 169, pp. 6515-6521.; Kitadai Y., Haruma K., Sumii K., Yamamoto S., Ue T., Yokozaki H., Yasui W., Ohmoto Y., Kajiyama G., Fidler I.J., Tahara E. Expression of IL-8 correlates with vascularity in human gastric carcinomas. Am. J. Pathol., 1998, vol. 152, pp. 93-100; K nig A., Krenn V., Toksoy A., Gerhard N., Gillitzer R. MIG, GROα and RANTES messenger RNA expression in lining layer, infiltrates and different leucocyte populations of synovial tissue from patients with rheumatoid arthritis, psoriatic arthritis and osteoarthritis. Virchows Arch., 2000, vol. 436, pp. 449-458.; Maurer A.M., Zhou B., Han Z.C. Roles of platelet factor 4 in hematopoiesis and angiogenesis. Growth Factors, 2006, vol. 24, pp. 242-252.; Murphy P.M., Baggiolini M., Charo I.F., Hebert C.A., Horuk R., Matsushima K., Miller L.H., Oppenheim J.J., Power C.A. International union of pharmacology. XXII. Nomenclature for chemokine receptors. Pharmacol. Rev., 2000, vol. 52, pp. 145-176.; Overbergh L., Giulietti A., Valckx D., Decallonne R., Bouillon R., Mathieu C. The use of real-time reverse transcriptase PCR for the quantification of cytokine gene expression. J. Biomol. Tech., 2003, vol. 14, no. 1, pp. 33-43.; Overbergh L, Gysemans C, Mathieu C. Quantification of chemokines by real-time reverse transcriptase PCR: applications in type 1 diabetes. Expert Rev. Mol. Diagn., 2006 Jan, vol. 6, no. 1, pp. 51-64.; Pierer M., Rethage J., Seibl R., Lauener R., Brentano F., Wagner U., Hantzschel H., Michel B.A., Gay R.E., Gay S., Kyburz D. Chemokine secretion of rheumatoid arthritis synovial fibroblasts stimulated by Tolllike receptor 2 ligands. J. Immunol., 2004, vol. 172, no. 2, pp. 1256-1265.; Schmittgen T., Livak K. Analyzing real-time PCR data by the comparative CT method. Nature Protocols, 2008, vol. 3, no. 6, pp. 1101-1108.; Schmutz C., Hulme A., Burman A., Salmon M., Ashton B., Buckley C., Middleton J. Chemokine receptors in the rheumatoid synovium: upregulation of CXCR5. Arthritis Res. Ther., 2005, vol. 7, no. 2, pp. R217-229.; Schroeder A., Mueller O., Stocker S., Salowsky R., Leiber M., Gassmann M., Lightfoot S., Menzel W., Granzow M., Ragg T. The RIN: an RNA integrity number for assigning integrity values to RNA measurements. BMC Mol Biol., 2006, vol. 7, p. 3 http://www.biomedcentral.com/1471-2199/7/3; Stamatovic S.M., Keep R.F., Mostarica-Stojkovic M., Andjelkovic A.V. CCL2 regulates angiogenesis via activation of Ets-1 transcription factor. J. Immunol., 2006, vol. 177, pp. 2651-2661.; Strieter R.M., Polverini P.J., Kunkel S.L., Arenberg D.A., Burdick M.D., Kasper J., Dzuiba J., Van Damme J., Walz A., Marriott D., Chan S.-Y., Roczniak S., Shanafelt A.B. The functional role of the ELR motif in CXC chemokine-mediated angiogenesis. J. Biol. Chem., 1995, vol. 270, pp. 27348-27357.; Van Damme J., Mantovani A. From cytokines to chemokines. Cytokine & Growth Factor Reviews 16, 2005, pp. 549-551.; Van der Voort R., Antoine W.T. van Lieshout, Liza W.J. Toonen, Annet W. Sl etjes, Wim B. van den Berg, Carl G. Figdor, Timothy R.D.J. Radstake, Gosse J. Adema. Elevated CXCL16 expression by synovial macrophages recruits memory T cells into rheumatoid joints. Arthritis Rheum., 2005, vol. 52, no. 5, pp. 1381-1391.; Vandesompele J., De Preter K., Pattyn F., Poppe B., Van Roy N., De Paepe A., Speleman F. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biology, 2002, vol. 3, no. 7: http://genomebiology.com/2002/3/7/research/0034.5; Volin M.V., Woods J.M., Amin M.A., Connors M.A., Harlow L.A., Koch A.E. Fractalkine: A novel angiogenic chemokine in rheumatoid arthritis. Am. J. Pathol., 2001, vol. 159, pp. 1521-1526.; Yoshida S., Arakawa F., Higuchi F., Ishibashi Y., Goto M., Sugita Y., Nomura Y., Niino D., Shimizu K., Aoki R., Hashikawa K., Kimura Y., Yasuda K., Tashiro K., Kuhara S., Nagata K., Ohshima K. Gene expression analysis of rheumatoid arthritis synovial lining regions by cDNA microarray combined with laser microdissection: up-regulation of inflammation-associated STAT1, IRF1, CXCL9, CXCL10, and CCL5. Scand. J. Rheumatol., 2012, vol. 41, pp. 170-179.; Zlotnik A., Yoshie O. Chemokines: a new classification system and their role in immunity. Immunity, 2000, vol. 12, pp. 121-127.; https://www.mimmun.ru/mimmun/article/view/676

  18. 18
    Academic Journal

    المصدر: Medical Immunology (Russia); Том 16, № 2 (2014); 189-194 ; Медицинская иммунология; Том 16, № 2 (2014); 189-194 ; 2313-741X ; 1563-0625 ; 10.15789/1563-0625-2014-2

    وصف الملف: application/pdf

    Relation: https://www.mimmun.ru/mimmun/article/view/685/688; Zhebrun D.A., Maslyanskiy A.L., Titov A.G., Patrukhin A.P., Kostareva A.A., Gol`tseva I.S, Totolian Areg A. Opredelenie mRNK angiogennykh i angiostaticheskikh khemokinov i ikh retseptorov v sinovial`noy obolochke metodom kolichestvennoy PTSR v real`nom vremeni [Analysis of expression of angiogenic and angiostatic chemokines and their receptors in synovial tissue by quantitative real-time PCR]. Meditsinskaya immunologiya – Medical Immunology, 2013, vol. 15, no. 6, pp. 525-534.; Beekhuizen M., Gierman L.M., Creemers L.B., Dhert W.J., Huizinga T.W, Van Osch G.J., Zuurmond A.M. A descriptive study of synovial fluid changes in cytokine, chemokine and growth factor levels between osteoarthritis patients and healthy donors. Osteoarthritis and Cartilage, 2012, vol. 20, s. 1, pp. S79-S80.; Beekhuizen M., Gierman L.M., van Spil W.E., Van Osch G.J., Huizinga T.W., Saris D.B., Creemers L.B., Zuurmond A.M. An explorative study comparing levels of soluble mediators in control and osteoarthritic synovial fluid. Osteoarthritis and Cartilage, 2013, vol. 21, pp. 918-922.; Blades M.C., Ingegnoli F., Wheller S.K., Manzo A., Wahid S, Panayi G.S., Perretti M., Pitzalis C. Stromal cell-derived factor 1 (CXCL12) induces monocyte migration into human synovium transplanted onto SCID Mice. Arthritis Rheum., 2002, vol. 46, no. 3, pp. 824-836.; De Jager W., Hoppenreijs E.P., Wulffraat N.M., Wedderburn L.R., Kuis W., Prakken B.J. Blood and synovial fluid cytokine signatures in patients with juvenile idiopathic arthritis: a cross-sectional study. Ann. Rheum. Dis., 2007, vol. 66, no. 5, pp. 589-598.; Hampel U., Sesselmann S., Iserovich P., Sel S., Paulsen F., Sack R. Chemokine and cytokine levels in osteoarthritis and rheumatoid arthritis synovial fluid. J. of Immunological Methods, 2013, vol. 396, pp. 134-139.; Hitchon C.A., Alex P., Erdile L.B., Frank M.B., Dozmorov I., Tang Y., Wong K., Centola M., El-Gabalawy H.S. A distinct multicytokine profile is associated with anti-cyclical citrullinated peptide antibodies in patients with early untreated inflammatory arthritis. J. Rheumatol., 2004, vol. 31, no. 12, pp. 2336-2346.; K nig A., Krenn V., Toksoy A., Gerhard N., Gillitzer R. MIG, GROa and RANTES messenger RNA expression in lining layer, infiltrates and different leucocyte populations of synovial tissue from patients with rheumatoid arthritis, psoriatic arthritis and osteoarthritis. Virchows Arch., 2000, vol. 436, pp. 449-458.; Pierer M., Rethage J., Seibl R., Lauener R., Brentano F., Wagner U., Hantzschel H., Michel B.A., Gay R.E., Gay S., Kyburz D. Chemokine secretion of rheumatoid arthritis synovial fibroblasts stimulated by Tolllike receptor 2 ligands. J. Immunol., 2004, vol. 172, no. 2, pp. 1256-1265.; Pierzchala A.W., Kusz D.J., Hajduk G. CXCL8 and CCL5 Expression in Synovial Fluid and Blood Serum in Patients with Osteoarthritis of the Knee. Arch. Immunol. Ther. Exp., 2011, vol. 59, no. 2, pp. 151-155.; Vangsness C.T. Jr., Burke W.S., Narvy S.J., MacPhee R.D., Fedenko A.N. Human knee synovial fluid cytokines correlated with grade of knee osteoarthritis-a pilot study. Bull. NYU Hosp. Jt. Dis., 2011, vol. 69, no. 2, pp. 122-127.; https://www.mimmun.ru/mimmun/article/view/685

  19. 19
    Academic Journal

    المصدر: Medical Immunology (Russia); Том 9, № 1 (2007); 15-34 ; Медицинская иммунология; Том 9, № 1 (2007); 15-34 ; 2313-741X ; 1563-0625 ; 10.15789/1563-0625-2007-1

    وصف الملف: application/pdf

    Relation: https://www.mimmun.ru/mimmun/article/view/116/117; Baecklund E., Askling J., Rosenquist R., Ekbom A., Klareskog L. Rheumatoid arthritis and malignant lymphomas // Curr. Opin. Rheumatol. – 2004. – Vol. 1, N 3. – P. 254-261.; Geborek P., Bladstrom A., Turesson C., Gulfe A., Petersson I.F., Saxne T., Olsson H., Jacobsson L.T. Tumour necrosis factor blockers do not increase overall tumour risk in patients with rheumatoid arthritis, but may be associated with an increased risk of lymphomas // Ann. Rheum. Dis. – 2005. – Vol. 64, N 5. – P. 699-703.; Listing J., Strangfeld A., Kary S., Rau R., von Hinueber U., Stoyanova-Scholz M., Gromnica-Ihle E., Antoni C., Herzer P., Kekow J., Schneider M., Zink A. Infections in patients with rheumatoid arthritis treated with biologic agents // Arthritis Rheum. – 2005. – Vol. 52, N 11. – P. 3403-3412.; Marmont A.M. Stem cell transplantation for severe autoimmune diseases: progress and problems // Haematologica. – 1998. – Vol. 83, N 8. – P. 733-743.; Lorenz H.M., Herrmann M., Kalden J.R. The pathogenesis of autoimmune diseases // Scand. J. Clin. Lab. Invest. Suppl. – 2001. – Vol. 235. – P. 16-26.; Panayi G.S., Corrigall V.M., Pitzalis C. Pathogenesis of rheumatoid arthritis. The role of T cells and other beasts // Rheum. Dis. Clin. North. Am. – 2001. – Vol. 27, N 2. – P. 317-334.; Weyand C.M., Bryl E., Goronzy J.J. The role of T-cells in rheumatoid arthritis // Arch. Immunol. Ther. Exp. (Warsz.). – 2000. – Vol. 48, N 5. – P. 429-35.; Kinne R.W., Brauer R., Stuhlmuller B., Palombo-Kinne E., Burmester G.R. Macrophages in rheumatoid arthritis // Arthritis Res. – 2000. – Vol. 2, N 3. – P. 189-202.; Imamura F., Aono H., Hasunuma T., Sumida T., Tateishi H., Maruo S., Nishioka K. Monoclonal expansion of synoviocytes in rheumatoid arthritis // Arthritis Rheum. – 1998. – Vol. 41, N 11. – P. 1979-1986.; Isaacs J.D., Burrows N., Wing M., Keogan M.T., Rebello P.R., Watts R.A., Pye R.J., Norris P., Hazelman B.L., Hale G., Waldmann H. Humanized anti-CD4 monoclonal antibody therapy of autoimmune and inflammatory disease // Clin. Exp. Immunol. – 1997. – Vol. 110, N 2. – P. 158-166.; Matteson E.L., Yocum D.E., St Clair E.W., Achkar A.A., Thakor M.S., Jacobs M.R., Hays A.E., Heitman C.K., Johnston J.M. Treatment of active refractory rheumatoid arthritis with humanized monoclonal antibody CAMPATH-1H administered by daily subcutaneous injection // Arthritis Rheum. – 1995. – Vol. 38, N 9. – P. 1187-1193.; Lee D.M., Friend D.S., Gurish M.F., Benoist C., Mathis D., Brenner M.B. Mast cells: a cellular link between autoantibodies and inflammatory arthritis // Science. – 2002. – Vol. 297, N 5587. – P. 1689-1692.; Lovgren T., Eloranta M.L., Bave U., Alm G.V., Ronnblom L. Induction of interferon-alpha production in plasmacytoid dendritic cells by immune complexes containing nucleic acid released by necrotic or late apoptotic cells and lupus IgG // Arthritis Rheum. – 2004. – Vol. 50, N 6. – P. 1861-1872.; Martin F., Chan A.C. B-cell immunobiology in disease: evolving concepts from the clinic // Annu. Rev. Immunol. – 2006. – Vol. 24. – P. 467-496.; Vallin H., Perers A., Alm G.V., Ronnblom L. Anti-double-stranded DNA antibodies and immunostimulatory plasmid DNA in combination mimic the endogenous IFN-alpha inducer in systemic lupus erythematosus // J. Immunol. – 1999. – Vol. 163, N 11. – P. 6306-6313.; Chan O.T., Hannum L.G., Haberman A.M., Madaio M.P., Shlomchik M.J. A novel mouse with B cells but lacking serum antibody reveals an antibodyindependent role for B cells in murine lupus // J. Exp. Med. – 1999. – Vol. 189, N 10. – P. 1639-1648.; Wong F.S., Wen L., Tang M., Ramanathan M., Visintin I., Daugherty J., Hannum L.G., Janeway Jr. C.A., Shlomchik M.J. Investigation of the role of B-cells in type 1 diabetes in the NOD mouse // Diabetes. – 2004. – Vol. 53, N 10. – P. 2581-2587.; O’Neill S.K., Shlomchik M.J., Glant T.T., Cao Y., Doodes P.D., Finnegan A. Antigen-specific B cells are required as APCs and autoantibody-producing cells for induction of severe autoimmune arthritis // J. Immunol. – 2005. – Vol. 174, N 6. – P. 3781-3788.; Soderstrom N.A.B. Organization of the invading lymphoid tissue in human lymphoid thyroiditis // Scand. J. Immunol. – 1974. – Vol. 3, N 3. – P. 295-301.; Prineas J.W. Multiple sclerosis: presence of lymphatic capillaries and lymphoid tissue in the brain and spinal cord // Science. – 1979. – Vol. 203, N 4385. – P. 1123-1125.; Stott D.I., Hiepe F., Hummel M., Steinhauser G., Berek C. Antigen-driven clonal proliferation of B cells within the target tissue of an autoimmune disease. The salivary glands of patients with Sjogren’s syndrome // J. Clin. Invest. – 1998. – Vol. 102, N 5. – P. 938-946.; Lorenz R.G., Chaplin D.D., McDonald K.G., McDonough J.S., Newberry R.D. Isolated lymphoid follicle formation is inducible and dependent upon lymphotoxin-sufficient B lymphocytes, lymphotoxin beta receptor, and TNF receptor I function // J. Immunol. – 2003. – Vol. 170, N 11. – P. 5475-5482.; Ansel K.M., Ngo V.N., Hyman P.L., Luther S.A., Forster R., Sedgwick J.D., Browning J.L., Lipp M., Cyster J.G. A chemokine-driven positive feedback loop organizes lymphoid follicles // Nature. – 2000. – Vol. 406, N 6793. – P. 309-314.; Mariathasan S., Matsumoto M., Baranyay F., Nahm M.H., Kanagawa O., Chaplin D.D. Absence of lymph nodes in lymphotoxin-alpha (LT alpha)-deficient mice is due to abnormal organ development, not defective lymphocyte migration // J. Inflamm. – 1995. – Vol. 45, N 1. – P. 72-78.; Matsumoto M., Mariathasan S., Nahm M.H., Baranyay F., Peschon J.J., Chaplin D.D. Role of lymphotoxin and the type I TNF receptor in the formation of germinal centers // Science. – 1996. – Vol. 271, N 5253. – P. 1289-1291.; Ngo V.N., Cornall R.J., Cyster J.G. Splenic T-zone development is B cell dependent // J. Exp. Med. – 2001. – Vol. 194, N 11. – P. 1649-1660.; Kurt-Jones E.A., Liano D., HayGlass K.A., Benacerraf B., Sy M.S., Abbas A.K. The role of antigen-presenting B cells in T cell priming in vivo. Studies of B cell-deficient mice // J. Immunol. – 1988. – Vol. 140, N 11. – P. 3773-3778.; Chan O., Shlomchik M.J. A new role for B cells in systemic autoimmunity: B cells promote spontaneous T cell activation in MRL-lpr/lpr mice // J. Immunol. – 1998. – Vol. 160, N 1. – P. 51-59.; Takemura S., Klimiuk P.A., Braun A., Goronzy J.J., Weyand C.M. T cell activation in rheumatoid synovium is B cell dependent // J. Immunol. – 2001. – Vol. 167, N 8. – P. 4710-4718.; Olson T.S., Bamias G., Naganuma M., Rivera-Nieves J., Burcin T.L., Ross W., Morris M.A., Pizarro T.T., Ernst P.B., Cominelli F., Ley K. Expanded B cell population blocks regulatory T cells and exacerbates ileitis in a murine model of Crohn disease // J. Clin Invest. – 2004. – Vol. 114, N 3. – P. 389-398.; Yu S., Maiti P.K., Dyson M., Jain R., Braley-Mullen H. B cell-deficient NOD.H-2h4 mice have CD4+CD25+ T regulatory cells that inhibit the development of spontaneous autoimmune thyroiditis // J. Exp. Med. – 2006. – Vol. 203, N 2. – P. 349-358.; Pistoia V. Production of cytokines by human B cells in health and disease // Immunol. Today. – 1997. – Vol. 18, N 7. – P. 343-350.; Mewar D., Wilson A.G. Autoantibodies in rheumatoid arthritis: a review // Biomed. Pharmacother. – 2006. – Vol. 60, N 10. – P. 648-655.; Nielen M.M., van Schaardenburg D., Reesink H.W., van de Stadt R.J., van der Horst-Bruinsma I.E., de Koning M.H., Habibuw M.R., Vandenbroucke J.P., Dijkmans B.A. Specific autoantibodies precede the symptoms of rheumatoid arthritis: a study of serial measurements in blood donors // Arthritis Rheum. – 2004. – Vol. 50, N 2. – P. 380-386.; Binder M., Otto F., Mertelsmann R., Veelken H., Trepel M. The epitope recognized by rituximab // Blood. – 2006. – Vol. 108, N 6. – P. 1975-1978.; Protheroe A., Edwards J.C., Simmons A., Maclennan K., Selby P. Remission of inflammatory arthropathy in association with anti-CD20 therapy for non-Hodgkin’s lymphoma // Rheumatology (Oxford). – 1999. – Vol. 38, N 11. – P. 1150-1152.; Leandro M.J., Edwards J.C., Cambridge G. Clinical outcome in 22 patients with rheumatoid arthritis treated with B-lymphocyte depletion // Ann. Rheum. Dis. – 2002. – Vol. 61, N 10. – P. 883-888.; Edwards J.C., Szczepanski L., Szechinski J., Filipowicz-Sosnowska A., Emery P., Close D.R., Stevens R.M., Shaw T. Efficacy of B-cell-targeted therapy with rituximab in patients with rheumatoid arthritis // N. Engl. J. Med. – 2004. – Vol. 350, N 25. – P. 2572-2581.; Emery P., Fleischmann R., Filipowicz-Sosnowska A., Schechtman J., Szczepanski L., Kavanaugh A., Racewicz A.J., van Vollenhoven R.F., Li N.F., Agarwal S., Hessey E.W., Shaw T.M. The efficacy and safety of rituximab in patients with active rheumatoid arthritis despite methotrexate treatment: results of a phase IIB randomized, double-blind, placebo-controlled, dose-ranging trial // Arthritis Rheum. – 2006. – Vol. 54, N 5. – P. 1390-1400.; Cohen S.B., Emery P., Greenwald M.W., Dougados M., Furie R.A., Genovese M.C., Keystone E.C., Loveless J.E., Burmester G.R., Cravets M.W., Hessey E.W., Shaw T., Totoritis M.C. Rituximab for rheumatoid arthritis refractory to anti-tumor necrosis factor therapy: Results of a multicenter, randomized, double-blind, placebo-controlled, phase III trial evaluating primary efficacy and safety at twenty-four weeks // Arthritis Rheum. – 2006. – Vol. 54, N 9. – P. 2793-2806.; Leandro M.J., Cambridge G., Edwards J.C., Ehrenstein M.R., Isenberg D.A. B-cell depletion in the treatment of patients with systemic lupus erythematosus: a longitudinal analysis of 24 patients // Rheumatology (Oxford). – 2005. – Vol. 44, N 12. – P. 1542-1545.; Anolik J.H., Barnard J., Cappione A., Pugh-Bernard A.E., Felgar R.E., Looney R.J., Sanz I. Rituximab improves peripheral B cell abnormalities in human systemic lupus erythematosus // Arthritis Rheum. – 2004. – Vol. 50, N 11. – P. 3580-3590.; Looney R.J., Anolik J.H., Campbell D., Felgar R.E., Young F., Arend L.J., Sloand J.A., Rosenblatt J., Sanz I. B cell depletion as a novel treatment for systemic lupus erythematosus: a phase I/II dose-escalation trial of rituximab // Arthritis Rheum. – 2004. – Vol. 50, N 8. – P. 2580-2589.; Pijpe J., van Imhoff G.W., Spijkervet F.K., Roodenburg J.L., Wolbink G.J., Mansour K., Vissink A., Kallenberg C.G., Bootsma H. Rituximab treatment in patients with primary Sjogren’s syndrome: an open-label phase II study // Arthritis Rheum. – 2005. – Vol. 52, N 9. – P. 2740-2750.; Keogh K.A., Wylam M.E., Stone J.H., Specks U. Induction of remission by B lymphocyte depletion in eleven patients with refractory antineutrophil cytoplasmic antibody-associated vasculitis // Arthritis Rheum. – 2005. – Vol. 52, N 1. – P. 262-268.; Levine T.D. Rituximab in the treatment of dermatomyositis: an open-label pilot study // Arthritis Rheum. – 2005. – Vol. 52, N 2. – P. 601-607.; Schmidt E., Hunzelmann N., Zillikens D., Brocker E.B., Goebeler M. Rituximab in refractory autoimmune bullous diseases // Clin. Exp. Dermatol. – 2006. – Vol. 31, N 4. – P. 503-508.; Hoffman P.C. Immune hemolytic anemia – selected topics // Hematology Am. Soc. Hematol. Educ. Program. – 2006. – P. 13-18.; Arnold D.M., Dentali F., Crowther M.A., Meyer R.M., Cook R.J., Sigouin C., Fraser G.A., Lim W., Kelton J.G. Systematic review: efficacy and safety of rituximab for adults with idiopathic thrombocytopenic purpura // Ann. Intern. Med. – 2007. – Vol. 146, N 1. – P. 25-33.; Bay A., Oner A.F., Uner A., Dogan M. Use of rituximab in chronic childhood immune thrombocytopenic purpura // Pediatr. Int. – 2006. – Vol. 48, N 5. – P. 514-516.; Kavanaugh A.F. B cell targeted therapies: safety considerations // J. Rheumatol. Suppl. – 2006. – Vol. 77. – P. 18-23.; De Vita S., Zaja F., Sacco S., De Candia A., Fanin R., Ferraccioli G. Efficacy of selective B cell blockade in the treatment of rheumatoid arthritis: evidence for a pathogenetic role of B cells // Arthritis Rheum. – 2002. – Vol. 46, N 8. – P. 2029-2033.; Kneitz C., Wilhelm M., Tony H.P. Improvement of refractory rheumatoid arthritis after depletion of B cells // Scand. J. Rheumatol. – 2004. – Vol. 33, N 2. – P. 82-86.; Moore J., Ma D., Will R., Cannell P., Handel M., Milliken S. A phase II study of Rituximab in rheumatoid arthritis patients with recurrent disease following haematopoietic stem cell transplantation // Bone Marrow Transplant. – 2004. – Vol. 34, N 3. – P. 241-247.; Roll P., Palanichamy A., Kneitz C., Dorner T., Tony H.P. Regeneration of B cell subsets after transient B cell depletion using anti-CD20 antibodies in rheumatoid arthritis // Arthritis Rheum. – 2006. – Vol. 54, N 8. – P. 2377-2386.; Petschner F., Walker U.A., Schmitt-Graff A., Uhl M., Peter H.H. «Catastrophic systemic lupus erythematosus» with Rosai-Dorfman sinus histiocytosis. Successful treatment with anti-CD20/ rutuximab // Dtsch. Med. Wochenschr. – 2001. – Vol. 126, N 37. – P. 998-1001.; Perrotta S., Locatelli F., La Manna A., Cennamo L., De Stefano P., Nobili B. Anti-CD20 monoclonal antibody (Rituximab) for life-threatening autoimmune haemolytic anaemia in a patient with systemic lupus erythematosus // Br. J. Haematol. – 2002. – Vol. 116, N 2. – P. 465-467.; Anolik J.H., Campbell D., Felgar R.E., Young F., Sanz I., Rosenblatt J., Looney R.J. The relationship of FcgammaRIIIa genotype to degree of B cell depletion by rituximab in the treatment of systemic lupus erythematosus // Arthritis Rheum. – 2003. – Vol. 48, N 2. – P. 455-459.; Weide R., Heymanns J., Pandorf A., Koppler H. Successful long-term treatment of systemic lupus erythematosus with rituximab maintenance therapy // Lupus. – 2003. – Vol. 12, N 10. – P. 779-782.; Tokunaga M., Fujii K., Saito K., Nakayamada S., Tsujimura S., Nawata M., Tanaka Y. Downregulation of CD40 and CD80 on B cells in patients with life-threatening systemic lupus erythematosus after successful treatment with rituximab // Rheumatology (Oxford). – 2005. – Vol. 44, N 2. – P. 176-182.; Edelbauer M., Jungraithmayr T., Zimmerhackl L.B. Rituximab in childhood systemic lupus erythematosus refractory to conventional immunosuppression: case report // Pediatr. Nephrol. – 2005. – Vol. 20, N 6. – P. 811-813.; van Vollenhoven R.F., Gunnarsson I., Welin-Henriksson E., Sundelin B., Osterborg A., Jacobson S.H., Klareskog L. Biopsy-verified response of severe lupus nephritis to treatment with rituximab (anti-D20 monoclonal antibody) plus cyclophosphamide after biopsy-documented failure to respond to cyclophosphamide alone // Scand. J. Rheumatol. – 2004. – Vol. 33, N 6. – P. 423-427.; Lambotte O., Durbach A., Kotb R., Ferlicot S., Delfraissy J.F., Goujard C. Failure of rituximab to treat a lupus flare-up with nephritis // Clin Nephrol. – 2005. – Vol. 64, N 1. – P. 73-77.; Sabugo F., Llanos C., Soto L., Gutierrez J., Cuchacovich M. Rituximab (anti-CD20 monoclonal antibody) for refractory systemic lupus erythematosus: report of one case // Rev. Med. Chil. – 2005. – Vol. 133, N 6. – P. 681-684.; Van den Bergh B., Selleslag D., Boelaert J.R., Matthys E.G., Schurgers M., Vandecasteele S., De Vriese A. Management of therapy-resistant systemic lupus erythematosus with rituximab: report of a case and review of the literature // Acta Clin. Belg. – 2005. – Vol. 60, N 2. – P. 102-105.; Marks S.D., Patey S., Brogan P.A., Hasson N., Pilkington C., Woo P., Tullus K. B lymphocyte depletion therapy in children with refractory systemic lupus erythematosus // Arthritis Rheum. – 2005. – Vol. 52, N 10. – P. 3168-3174.; Armstrong D., Wright S., McVeigh C., Finch M. Infective endocarditis complicating rituximab (anti-CD20 monoclonal antibody) treatment in an SLE patient with a past history of Libman-Sacks endocarditis: a case for antibiotic prophylaxis? // Clin. Rheumatol. – 2006. – Vol. 25, N 4. – P. 583-584.; Lehembre S., Macario-Barrel A., Musette P., Carvalho P., Joly P. Rituximab treatment for immune thrombocytopenia associated with systemic lupus erythematosus // Ann. Dermatol. Venereol. – 2006. – Vol. 133, N 1. – P. 53-55.; Lim S.W., Gillis D., Smith W., Hissaria P., Greville H., Peh C.A. Rituximab use in systemic lupus erythematosus pneumonitis and a review of current reports // Intern. Med. J. – 2006. – Vol. 36, N 4. – P. 260-262.; Gomard-Mennesson E., Ruivard M., Koenig M., Woods A., Magy N., Ninet J., Rousset H., Salles G., Broussolle C., Seve P. Treatment of isolated severe immune hemolytic anaemia associated with systemic lupus erythematosus: 26 cases // Lupus. – 2006. – Vol. 15, N 4. – P. 223-231.; Willems M., Haddad E., Niaudet P., Kone-Paut I., Bensman A., Cochat P., Deschenes G., Fakhouri F., Leblanc T., Llanas B., Loirat C., Pillet P., Ranchin B., Salomon R., Ulinski T., Bader-Meunier B. Rituximab therapy for childhood-onset systemic lupus erythematosus // J. Pediatr. – 2006. – Vol. 148, N 5. – P. 623-627.; Jansson A.F., U. Wintergerst, E.D. Renner, B.H. Belohradsky. Rituximab-induced long-term remission in two children with SLE // Eur. J. Pediatr. – 2007. – Vol. 166, N 2. – P. 177-181.; Smith K.G., Jones R.B., Burns S.M., Jayne D.R. Long-term comparison of rituximab treatment for refractory systemic lupus erythematosus and vasculitis: Remission, relapse, and retreatment // Arthritis Rheum. – 2006. – Vol. 54, N 9. – P. 2970-2982.; Tokunaga M., Saito K., Kawabata D., Imura Y., Fujii T., Nakayamada S., Tsujimura S., Nawata M., Iwata S., Azuma T., Mimori T., Tanaka Y. Efficacy of rituximab (Anti-CD20) for refractory systemic lupus erythematosus involving the central nervous system // Ann. Rheum. Dis., 2006.; Kotani T., Takeuchi T., Kawasaki Y., Hirano S., Tabushi Y., Kagitani M., Makino S., Hanafusa T. Successful treatment of cold agglutinin disease with anti-CD20 antibody (rituximab) in a patient with systemic lupus erythematosus // Lupus. – 2006. – Vol. 15, N 10. – P. 683-685.; Specks U., Fervenza F.C., McDonald T.J., Hogan M.C. Response of Wegener’s granulomatosis to anti-CD20 chimeric monoclonal antibody therapy // Arthritis Rheum. – 2001. – Vol. 44, N 12. – P. 2836-2840.; Eriksson P. Nine patients with anti-neutrophil cytoplasmic antibody-positive vasculitis successfully treated with rituximab // J. Intern. Med. – 2005. – Vol. 257, N 6. – P. 540-548.; Omdal R., Wildhagen K., Hansen T., Gunnarsson R., Kristoffersen G. Anti-CD20 therapy of treatment-resistant Wegener’s granulomatosis: favourable but temporary response // Scand. J. Rheumatol. – 2005. – Vol. 34, N 3. – P. 229-232.; Keogh K.A., Ytterberg S.R., Fervenza F.C., Carlson K.A., Schroeder D.R., Specks U. Rituximab for refractory Wegener’s granulomatosis: report of a prospective, open-label pilot trial // Am. J. Respir. Crit. Care Med. – 2006. – Vol. 173, N 2. – P. 180-187.; Aries P.M., Hellmich B., Voswinkel J., Both M., Nolle B., Holl-Ulrich K., Lamprecht P., Gross W.L. Lack of efficacy of rituximab in Wegener’s granulomatosis with refractory granulomatous manifestations // Ann. Rheum. Dis. – 2006. – Vol. 65, N 7. – P. 853-858.; Clatworthy M.R., Jayne D.R. Acquired hemophilia in association with ANCA-associated vasculitis: response to rituximab // Am. J. Kidney Dis. – 2006. – Vol. 47, N 4. – P. 680-682.; Stasi R., Stipa E., Del Poeta G., Amadori S., Newland A.C., Provan D. Long-term observation of patients with anti-neutrophil cytoplasmic antibodyassociated vasculitis treated with rituximab // Rheumatology (Oxford). – 2006. – Vol. 45, N 11. – P. 1432-1436.; Tektonidou M.G., Skopouli F.N. Sustained 3-year remission after rituximab treatment in a patient with refractory Wegener’s granulomatosis // Clin. Exp. Rheumatol. – 2006. – Vol. 24, N 2, Suppl. 41. – P. S103.; Koukoulaki M., Smith K.G., Jayne D.R. Rituximab in Churg-Strauss syndrome // Ann. Rheum. Dis. – 2006. – Vol. 65, N 4. – P. 557-559.; Somer B.G., Tsai D.E., Downs L., Weinstein B., Schuster S.J. Improvement in Sjogren’s syndrome following therapy with rituximab for marginal zone lymphoma // Arthritis Rheum. – 2003. – Vol. 49, N 3. – P. 394-398.; Seror R., Sordet C., Guillevin L., Hachulla E., Masson C., Ittah M., Candon S., Leguern V., Aouba A., Jean S., Gottenberg J.E., Mariette X. Tolerance and efficacy of rituximab and changes in serum B cell biomarkers in patients with systemic complications of primary Sjogren’s syndrome // Ann. Rheum. Dis., 2006.; Chiappetta N., Steier J., Gruber B. Rituximab in the treatment of refractory dermatomyositis // J. Clin. Rheumatol. – 2005. – Vol. 11, N 5. – P. 264-266.; Noss E.H., Hausner-Sypek D.L., Weinblatt M.E. Rituximab as therapy for refractory polymyositis and dermatomyositis // J. Rheumatol. – 2006. – Vol. 33, N 5. – P. 1021-1026.; Zaja F., Russo D., Fuga G., Patriarca F., Ermacora A., Baccarani M. Rituximab for the treatment of type II mixed cryoglobulinemia // Haematologica. – 1999. – Vol. 84, N 12. – P. 1157-1158.; Arzoo K., Sadeghi S., Liebman H.A. Treatment of refractory antibody mediated autoimmune disorders with an anti-CD20 monoclonal antibody (rituximab) // Ann. Rheum. Dis. – 2002. – Vol. 61, N 10. – P. 922-924.; Sansonno D., De Re V., Lauletta G., Tucci F.A., Boiocchi M., Dammacco F. Monoclonal antibody treatment of mixed cryoglobulinemia resistant to interferon alpha with an anti-CD20 // Blood. – 2003. – Vol. 101, N 10. – P. 3818-3826.; Lamprecht P., Lerin-Lozano C., Merz H., Dennin R.H., Gause A., Voswinkel J., Peters S.O., Gutzeit O., Arlt A.C., Solbach W., Gross W.L. Rituximab induces remission in refractory HCV associated cryoglobulinaemic vasculitis // Ann. Rheum. Dis. – 2003. – Vol. 62, N 12. – P. 1230-1233.; Roccatello D., Baldovino S., Rossi D., Mansouri M., Naretto C., Gennaro M., Cavallo R., Alpa M., Costanzo P., Giachino O., Mazzucco G., Sena L.M. Long-term effects of anti-CD20 monoclonal antibody treatment of cryoglobulinaemic glomerulonephritis // Nephrol. Dial. Transplant. – 2004. – Vol. 19, N 12. – P. 3054-3061.; Catuogno M., Rezai S., Priori R., Magrini L., Valesini G. Serum sickness associated with rituximab in a patient with hepatitis C virus-related mixed cryoglobulinaemia // Rheumatology (Oxford). – 2005. – Vol. 44, N 3. – P. 406.; Kay J., McCluskey R.T. Case records of the Massachusetts General Hospital. Case 31-2005. A 60-year-old man with skin lesions and renal insufficiency // N. Engl. J. Med. – 2005. – Vol. 353, N 15. – P. 1605-1613.; Cai F.Z., Ahern M., Smith M. Treatment of cryoglobulinemia associated peripheral neuropathy with rituximab // J. Rheumatol. – 2006. – Vol. 33, N 6. – P. 1197-1198.; Pekow J., Chung R.T. Treatment of type II cryoglobulinemia associated with hepatitis C with rituximab // J. Clin. Gastroenterol. – 2006. – Vol. 40, N 5. – P. 450.; Basse G., Ribes D., Kamar N., Mehrenberger M., Sallusto F., Esposito L., Guitard J., Lavayssiere L., Oksman F., Durand D., Rostaing L. Rituximab therapy for mixed cryoglobulinemia in seven renal transplant patients // Transplant. Proc. – 2006. – Vol. 38, N 7. – P. 2308-2310.; Cohen H., Green S., Jones S., Amos N., William B.D. Lack of efficacy of Rituximab in a patient with essential mixed cryoglobulinaemia // Rheumatology (Oxford), 2006.; Rubenstein E., Arkfeld D.G., Metyas S., Shinada S., Ehresmann S., Liebman H.A. Rituximab treatment for resistant antiphospholipid syndrome // J. Rheumatol. – 2006. – Vol. 33, N 2. – P. 355-357.; Anandacoomarasamy A., Gibson J., McGill N. «Cure» of life-threatening antiphospholipid syndrome with rituximab // Intern. Med. J. – 2006. – Vol. 36, N 7. – P. 474-475.; Ames P.R., Tommasino C., Fossati G., Scenna G., Brancaccio V., Ferrara F. Limited effect of rituximab on thrombocytopaenia and anticardiolipin antibodies in a patient with primary antiphospholipid syndrome // Ann. Hematol., 2006.; Ahn E.R., Lander G., Bidot C.J., Jy W., Ahn Y.S. Long-term remission from life-threatening hypercoagulable state associated with lupus anticoagulant (LA) following rituximab therapy // Am. J. Hematol. – 2005. – Vol. 78, N 2. – P. 127-129.; Hongeng S., Tardtong P., Worapongpaiboon S., Ungkanont A., Jootar S. Successful treatment of refractory autoimmune haemolytic anaemia in a post-unrelated bone marrow transplant paediatric patient with rituximab // Bone Marrow Transplant. – 2002. – Vol. 29, N 10. – P. 871-872.; Gupta N., Kavuru S., Patel D., Janson D., Driscoll N., Ahmed S., Rai K.R. Rituximab-based chemotherapy for steroid-refractory autoimmune hemolytic anemia of chronic lymphocytic leukemia // Leukemia. – 2002. – Vol. 16, N 10. – P. 2092-2095.; Zecca M., Nobili B., Ramenghi U., Perrotta S., Amendola G., Rosito P., Jankovic M., Pierani P., De Stefano P., Bonora M.R., Locatelli F. Rituximab for the treatment of refractory autoimmune hemolytic anemia in children // Blood. – 2003. – Vol. 101, N 10. – P. 3857-3861.; Motto D.G., Williams J.A., Boxer L.A. Rituximab for refractory childhood autoimmune hemolytic anemia // Isr. Med. Assoc. J. – 2002. – Vol. 4, N 11. – P. 1006-1008.; Gottardo N.G., Baker D.L., Willis F.R. Successful induction and maintenance of long-term remission in a child with chronic relapsing autoimmune hemolytic anemia using rituximab // Pediatr. Hematol. Oncol. – 2003. – Vol. 20, N 7. – P. 557-561.; Endo T., Nakao S., Koizumi K., Nishio M., Fujimoto K., Sakai T., Kumano K., Obara M., Koike T. Successful treatment with rituximab for autoimmune hemolytic anemia concomitant with proliferation of Epstein-Barr virus and monoclonal gammopathy in a post-nonmyeloablative stem cell transplant patient // Ann. Hematol. – 2004. – Vol. 83, N 2. – P. 114-116.; Shanafelt T.D., Madueme H.L., Wolf R.C., Tefferi A. Rituximab for immune cytopenia in adults: idiopathic thrombocytopenic purpura, autoimmune hemolytic anemia, and Evans syndrome // Mayo Clin. Proc. – 2003. – Vol. 78, N 11. – P. 1340-1346.; Raj A., Bertolone S., Cheerva A. Successful treatment of refractory autoimmune hemolytic anemia with monthly rituximab following nonmyeloablative stem cell transplantation for sickle cell disease // J. Pediatr. Hematol. Oncol. – 2004. – Vol. 26, N 5. – P. 312-314.; Erdozain J.G., Ruiz-Irastorza G., Egurbide M.V., Aguirre C. Sustained response to rituximab of autoimmune hemolytic anemia associated with antiphospholipid syndrome // Haematologica. – 2004. – Vol. 89, N 9. – P. ECR34.; Narat S., Gandla J., Hoffbrand A.V., Hughes R.G., Mehta A.B. Rituximab in the treatment of refractory autoimmune cytopenias in adults // Haematologica. – 2005. – Vol. 90, N 9. – P. 1273-1274.; van Daalen S.T., van Dijken P.J., Tamminga R.Y., Brons P.P. Rituximab instead of splenectomy in 4 children with chronic or refractory autoimmune haemolytic anaemia // Ned. Tijdschr. Geneeskd. – 2005. – Vol. 149, N 42. – P. 2350-2354.; Nishida H., Murase T., Ueno H., Park J.W., Yano T., Ikeda Y. Fludarabine-associated autoimmune hemolytic anemia occurring in B-cell chronic lymphocytic leukemia // Leuk. Res. – 2006. – Vol. 30, N 12. – P. 1589-1590.; Pamuk G.E., Turgut B., Demir M., Tezcan F., Vural O. The successful treatment of refractory autoimmune hemolytic anemia with rituximab in a patient with chronic lymphocytic leukemia // Am. J. Hematol. – 2006. – Vol. 81, N 8. – P. 631- 633.; Saleh M.N., Gutheil J., Moore M., Bunch P.W., Butler J., Kunkel L., Grillo-Lopez A.J., LoBuglio A.F. A pilot study of the anti-CD20 monoclonal antibody rituximab in patients with refractory immune thrombocytopenia // Semin. Oncol. – 2000. – Vol. 27, N 6, Suppl. 12. – P. 99-103.; Shvidel L., Klepfish A., Berrebi A. Successful treatment with Rituximab for relapsing immune thrombocytopenic purpura (ITP) associated with lowgrade non-Hodgkin’s lymphoma // Am. J. Hematol. – 2001. – Vol. 67, N 3. – P. 213-214.; Faurschou M., Hasselbalch H.C., Nielsen O.J. Sustained remission of platelet counts following monoclonal anti-CD20 antibody therapy in two cases of idiopathic autoimmune thrombocytopenia and neutropenia // Eur. J. Haematol. – 2001. – Vol. 66, N 6. – P. 408-411.; Stasi R., Pagano A., Stipa E., Amadori S. Rituximab chimeric anti-CD20 monoclonal antibody treatment for adults with chronic idiopathic thrombocytopenic purpura // Blood. – 2001. – Vol. 98, N 4. – P. 952-957.; Abdel-Raheem M.M., Potti A., Kobrinsky N. Severe Evans’s syndrome secondary to interleukin-2 therapy: treatment with chimeric monoclonal anti-CD20 antibody // Ann. Hematol. – 2001. – Vol. 80, N 9. – P. 543-545.; Zaja F., Iacona I., Masolini P., Russo D., Sperotto A., Prosdocimo S., Patriarca F., de Vita S., Regazzi M., Baccarani M., Fanin R. B-cell depletion with rituximab as treatment for immune hemolytic anemia and chronic thrombocytopenia // Haematologica. – 2002. – Vol. 87, N 2. – P. 189-195.; Delgado J., Bustos J.G., Jimenez-Yuste V., Hernandez-Navarro F. Anti-CD20 monoclonal antibody therapy in refractory immune thrombocytopenic purpura // Haematologica. – 2002. – Vol. 87, N 2. – P. 215-216.; Cohen Y., Polliack A. Sustained complete remission of chronic refractory immune thrombocytopenic purpura (ITP) of 10 years duration after only two infusions of rituximab // Hematol. J. – 2002. – Vol. 3, N 1. – P. 61-62.; Meo P., Stipa E., La Presa M., Bianchi M., Di Giulio C., Stasi R., Amadori S. Rituximab treatment of chronic idiopathic thrombocytopenic purpura. Results of a phase II study // Recenti Prog. Med. – 2002. – Vol. 93, N 7-8. – P. 421-427.; Giagounidis A.A., Anhuf J., Schneider P., Germing U., Sohngen D., Quabeck K., Aul C. Treatment of relapsed idiopathic thrombocytopenic purpura with the anti-CD20 monoclonal antibody rituximab: a pilot study // Eur. J. Haematol. – 2002. – Vol. 69, N 2. – P. 95-100.; D’Arena G., Luigiavigliotti M., Coccaro M., Iodice G., Tartarone A., Matera R., Di Renzo N. Late and long-lasting response in an adult chronic idiopathic thrombocytopenic purpura after extended course of rituximab // Leuk. Lymphoma. – 2003. – Vol. 44, N 3. – P. 561-562.; Riksen N.P., Keuning J.J., Vreugdenhil G. Rituximab in the treatment of relapsing idiopathic thrombocytopenic purpura // Neth. J. Med. – 2003. – Vol. 61, N 7. – P. 262-265.; Bengtson K.L., Skinner M.A., Ware R.E. Successful use of anti-CD20 (rituximab) in severe, lifethreatening childhood immune thrombocytopenic purpura // J. Pediatr. – 2003. – Vol. 143, N 5. – P. 670-673.; Thude H., Gruhn B., Werner U., Schorner U., Hafer R., Zintl F., Barz D. Treatment of a patient with chronic immune thrombocytopenic purpura with rituximab and monitoring by flow cytometric analysis // Acta Haematol. – 2004. – Vol. 111, N 4. – P. 221-224.; Cooper N., Stasi R., Cunningham-Rundles S., Feuerstein M.A., Leonard J.P., Amadori S., Bussel J.B. The efficacy and safety of B-cell depletion with anti-CD20 monoclonal antibody in adults with chronic immune thrombocytopenic purpura // Br. J. Haematol. – 2004. – Vol. 125, N 2. – P. 232-239.; Pusiol A., Cesaro S., Nocerino A., Picco G., Zanesco L., Bisogno G. Successful treatment with the monoclonal antibody rituximab in two children with refractory autoimmune thrombocytopenia // Eur. J. Pediatr. – 2004. – Vol. 163, N 6. – P. 305-307.; Koulova L., Alexandrescu D., Dutcher J.P., O’Boyle K.P., Eapen S., Wiernik P.H. Rituximab for the treatment of refractory idiopathic thrombocytopenic purpura (ITP) and thrombotic thrombocytopenic purpura (TTP): report of three cases // Am. J. Hematol. – 2005. – Vol. 78, N 1. – P. 49-54.; Tanai C., Iki S., Nakahara F., Iijima K., Usuki K., Kuwana M., Urabe A. Effective treatment with rituximab in a patient with refractory idiopathic thrombocytopenic purpura // Rinsho Ketsueki. – 2004. – Vol. 45, N 11. – P. 1181-1186.; Lalayanni C., Stavroyianni N., Saloum R., Tsompanakou A., Anagnostopoulos A. Rituximab is effective for selected patients with chronic steroidrefractory immune thrombocytopenic purpura // Hematology. – 2004. – Vol. 9, N 4. – P. 287-289.; Taube T., Schmid H., Reinhard H., von Stackelberg A., Overberg U.S. Effect of a single dose of rituximab in chronic immune thrombocytopenic purpura in childhood // Haematologica. – 2005. – Vol. 90, N 2. – P. 281-283.; Braendstrup P., Bjerrum O.W., Nielsen O.J., Jensen B.A., Clausen N.T., Hansen P.B., Andersen I., Schmidt K., Andersen T.M., Peterslund N.A., Birgens H.S., Plesner T., Pedersen B.B., Hasselbalch H.C. Rituximab chimeric anti-CD20 monoclonal antibody treatment for adult refractory idiopathic thrombocytopenic purpura // Am. J. Hematol. – 2005. – Vol. 78, N 4. – P. 275-280.; Moschovi M., Trimis G., Pergantou H., Platokouki H., Vrachnou E., Tzortzatou-Stathopoulou F. Clinical remission following monoclonal anti-CD20 therapy in two children with chronic refractory idiopathic thrombocytopenic purpura // J. Paediatr. Child Health. – 2005. – Vol. 41, N 7. – P. 384-386.; Carbone J., Escudero A., Mayayo M., Ballesteros M., Perez-Corral A., Sanchez-Ramon S., Sarmiento E., Micheloud D., Fernandez-Cruz E. Partial response to anti-CD20 monoclonal antibody treatment of severe immune thrombocytopenic purpura in a patient with common variable immunodeficiency. // Ann. N. Y. Acad. Sci. – 2005. – Vol. 1051. – P. 666-671.; Bennett C.M., Rogers Z.R., Kinnamon D.D., Bussel J.B., Mahoney D.H., Abshire T.C., Sawaf H., Moore T.B., Loh M.L., Glader B.E., McCarthy M.C., Mueller B.U., Olson T.A., Lorenzana A.N., Mentzer W.C., Buchanan G.R., Feldman H.A., Neufeld E.J. Prospective phase 1/2 study of rituximab in childhood and adolescent chronic immune thrombocytopenic purpura // Blood. – 2006. – Vol. 107, N 7. – P. 2639-2642.; Ojeda-Uribe M., Brunot A., Issler M. Successful treatment of idiopathic acquired refractory thrombotic thrombocytopenic purpura with an association of rituximab-vindesine. Report of one case // Rev. Med. Chil. – 2005. – Vol. 133, N 11. – P. 1349-1354.; Penalver F.J., Jimenez-Yuste V., Almagro M., Alvarez-Larran A., Rodriguez L., Casado M., Gallur L., Giraldo P., Hernandez R., Menor D., Rodriguez M.J., Caballero D., Gonzalez R., Mayans J., Millan I., Cabrera J.R. Rituximab in the management of chronic immune thrombocytopenic purpura: an effective and safe therapeutic alternative in refractory patients // Ann. Hematol. – 2006. – Vol. 85, N 6. – P. 400-406.; Taylor R.M., Bockenstedt P., Su G.L. Marrero J.A., Pellitier S.M., Fontana R.J. Immune thrombocytopenic purpura following liver transplantation: a case series and review of the literature // Liver Transpl. – 2006. – Vol. 12, N 5. – P. 781-791.; Parodi E., Nobili B., Perrotta S., Rosaria Matarese S.M., Russo G., Licciardello M., Zecca M., Locatelli F., Cesaro S., Bisogno G., Giordano P., De Mattia D., Ramenghia U. Rituximab (anti-CD20 monoclonal antibody) in children with chronic refractory symptomatic immune thrombocytopenic purpura: efficacy and safety of treatment // Int. J. Hematol. – 2006. – Vol. 84, N 1. – P. 48-53.; Latifzadeh S.Z., Entezari V. Chronic refractory idiopathic thrombocytopenic purpura (ITP) and anti-CD20 monoclonal antibody: a case report // Clin. Appl. Thromb. Hemost. – 2006. – Vol. 12, N 4. – P. 489-492.; Wiestner, A., Cho H.J., Asch A.S., Michelis M.A., Zeller J.A., Peerschke E.I., Weksler B.B., Schechter G.P. Rituximab in the treatment of acquired factor VIII inhibitors // Blood. – 2002. – Vol. 100, N 9. – P. 3426-3428.; Kain S., Copeland T.S., LeahyM.F. Treatment of refractory autoimmune (acquired) haemophilia with anti-CD20 (rituximab) // Br. J. Haematol. – 2002. – Vol. 119, N 2. – P. 578.; Marietta M., Pozzi S., Luppi M., Bertesi M., Cappi C., Morselli M., Torelli G. Acquired haemophilia in HIV negative, HHV-8 positive multicentric Castleman’s disease: a case report // Eur. J. Haematol. – 2003. – Vol. 70, N 3. – P. 181-182.; Stasi R., Brunetti M., Stipa E., Amadori S. Selective B-cell depl etion with rituximab for the treatment of patients with acquired hemophilia // Blood. – 2004. – Vol. 103, N 12. – P. 4424-4428.; Huang Y.W., Saidi P., Philipp C. Acquired factor VIII inhibitors in non-haemophilic patients: clinical experience of 15 cases // Haemophilia. – 2004. – Vol. 10, N 6. – P. 713-721.; Holme P.A., Brosstad F., Tjonnfjord G.E. Acquired haemophilia: management of bleeds and immune therapy to eradicate autoantibodies // Haemophilia. – 2005. – Vol. 11, N 5. – P. 510-515.; Abdallah A., Coghlan D.W., Duncan E.M., Chunilal S.D., Lloyd J.V. Rituximab-induced longterm remission in patients with refractory acquired hemophilia // J. Thromb. Haemost. – 2005. – Vol. 3, N 11. – P. 2589-2590.; Berezne A., Stieltjes N., Le-Guern V., Teixeira L., Billy C., Roussel-Robert V., Flaujac C., Horellou M.H., Guillevin L., Mouthon L. Rituximab alone or in association with corticosteroids in the treatment of acquired factor VIII inhibitors: report of two cases // Transfus. Med. – 2006. – Vol. 16, N 3. – P. 209-212.; Rojas-Garcia R., Gallardo E., de Andres I., de Luna N., Juarez C., Sanchez P., Illa I. Chronic neuropathy with IgM anti-ganglioside antibodies: lack of long term response to rituximab // Neurology. – 2003. – Vol. 61, N 12. – P. 1814-1816.; Ruegg S.J., Fuhr P., Steck A.J. Rituximab stabilizes multifocal motor neuropathy increasingly less responsive to IVIg // Neurology. – 2004. – Vol. 63, N 11. – P. 2178-2179.; Goldfarb A.R., Weimer L.H., Brannagan, 3rd T.H. Rituximab treatment of an IgM monoclonal autonomic and sensory neuropathy // Muscle Nerve. – 2005. – Vol. 31, N 4. – P. 510-515.; Broglio L., Lauria G. Worsening after rituximab treatment in anti-mag neuropathy // Muscle Nerve. – 2005. – Vol. 32, N 3. – P. 378-379.; Kilidireas C., Anagnostopoulos A., Karandreas N., Mouselimi L., Dimopoulos M.A. Rituximab therapy in monoclonal IgM-related neuropathies // Leuk. Lymphoma. – 2006. – Vol. 47, N 5. – P. 859-864.; Kelly J.J. Chronic peripheral neuropathy responsive to rituximab // Rev. Neurol. Dis. – 2006. – Vol. 3, N 2. – P. 78-81.; Gorson K.C., Natarajan N., Ropper A.H., Weinstein R. Rituximab treatment in patients with IVIg-dependent immune polyneuropathy: A prospective pilot trial // Muscle Nerve. – 2007. – Vol. 35, N 1. – P. 66-69.; Zaja F., Russo D., Fuga G., Perella G., Baccarani M. Rituximab for myasthenia gravis developing after bone marrow transplant // Neurology. – 2000. – Vol. 55, N 7. – P. 1062-1063.; Wylam M.E., Anderson P.M., Kuntz N.L., Rodriguez V. Successful treatment of refractory myasthenia gravis using rituximab: a pediatric case report // J. Pediatr. – 2003. – Vol. 143, N 5. – P. 674-677.; Gajra A., Vajpayee N., Grethlein S.J. Response of myasthenia gravis to rituximab in a patient with non-Hodgkin lymphoma // Am. J. Hematol. – 2004. – Vol. 77, N 2. – P. 196-197.; Takagi K., Yoshida A., Iwasaki H., Inoue H., Ueda T. Anti-CD20 antibody (Rituximab) therapy in a myasthenia gravis patient with follicular lymphoma // Ann. Hematol. – 2005. – Vol. 84, N 8. – P. 548-550.; Hain B., Jordan K., Deschauer M., Zierz S. Successful treatment of MuSK antibody-positive myasthenia gravis with rituximab // Muscle Nerve. – 2006. – Vol. 33, N 4. – P. 575-580.; Borradori L., Lombardi T., Samson J., Girardet C., Saurat J.H., Hugli A. Anti-CD20 monoclonal antibody (rituximab) for refractory erosive stomatitis secondary to CD20(+) follicular lymphoma-associated paraneoplastic pemphigus // Arch. Dermatol. – 2001. – Vol. 137, N 3. – P. 269-272.; Salopek T.G., Logsetty S., Tredget E.E. Anti-CD20 chimeric monoclonal anti-body (rituximab) for the treatment of recalcitrant, life-threatening pemphigus vulgaris with implications in the pathogenesis of the disorder // J. Am. Acad. Dermatol. – 2002. – Vol. 47, N 5. – P. 785-788.; Herrmann G., Hunzelmann N., Engert A. Treatment of pemphigus vulgaris with anti-CD20 monoclonal antibody (rituximab) // Br. J. Dermatol. – 2003. – Vol. 148, N 3. – P. 602-603.; Cooper H.L., Healy E., Theaker J.M., Friedmann P.S. Treatment of resistant pemphigus vulgaris with an anti-CD20 monoclonal antibody (Rituximab) // Clin. Exp. Dermatol. – 2003. – Vol. 28, N 4. – P. 366-368.; Goebeler M., Herzog S., Brocker E.B., Zillikens D. Rapid response of treatment-resistant pemphigus foliaceus to the anti-CD20 antibody rituximab // Br. J. Dermatol. – 2003. – Vol. 149, N 4. – P. 899-901.; Dupuy A., Viguier M., Bedane C., Cordoliani F., Blaise S., Aucouturier F., Bonnetblanc J.M., Morel P., Dubertret L., Bachelez H. Treatment of refractory pemphigus vulgaris with rituximab (anti-CD20 monoclonal antibody) // Arch. Dermatol. – 2004. – Vol. 140, N 1. – P. 91-96.; Espana A., Fernandez-Galar M., Lloret P., Sanchez-Ibarrola A., Panizo C. Long-term complete remission of severe pemphigus vulgaris with monoclonal anti-CD20 antibody therapy and immunophenotype correlations // J. Am. Acad. Dermatol. – 2004. – Vol. 50, N 6. – P. 974-976.; Morrison L.H. Therapy of refractory pemphigus vulgaris with monoclonal anti-CD20 antibody (rituximab) // J. Am. Acad. Dermatol. – 2004. – Vol. 51, N 5. – P. 817-819.; Wenzel J., Bauer R., Bieber T., Tuting T. Successful rituximab treatment of severe pemphigus vulgaris resistant to multiple immunosuppressants // Acta Derm. Venereol. – 2005. – Vol. 85, N 2. – P. 185-186.; Arin M.J., Engert A., Krieg T., Hunzelmann N. Anti-CD20 monoclonal antibody (rituximab) in the treatment of pemphigus // Br. J. Dermatol. – 2005. – Vol. 153, N 3. – P. 620-625.; Kong H.H., Prose N.S., Ware R.E., Hall, 3rd, R.P. Successful treatment of refractory childhood pemphgus vulgaris with anti-CD20 monoclonal antibody (rituximab) // Pediatr. Dermatol. – 2005. – Vol. 22, N 5. – P. 461-464.; Cecchi R., Gasperini U. Severe pemphigus vulgaris treated with rituximab (Mabthera). J. Dermatol. – 2005. – Vol. 32, N 10. – P. 862-864.; Esposito M., Capriotti E., Giunta A., Bianchi L., Chimenti S. Long-lasting remission of pemphigus vulgaris treated with rituximab // Acta Derm. Venereol. – 2006. – Vol. 86, N 1. – P. 87-89.; Niedermeier A., Worl P., Barth S., Schuler G., Hertl M. Delayed response of oral pemphigus vulgaris to rituximab treatment // Eur. J. Dermatol. – 2006. – Vol. 16, N 3. – P. 266-270.; Borel C., Launay F., Garrouste C., Astudillo L., Bazex J., Arlet P., Paul C., Viraben R., Sailler L. Rituximab induced remission of pemphigus vulgaris: 2 cases // Rev. Med. Interne, 2006.; Ahmed A.R., Spigelman Z., Cavacini L.A., Posner M.R. Treatment of pemphigus vulgaris with rituximab and intravenous immune globulin // N. Engl. J. Med. – 2006. – Vol. 355, N 17. – P. 1772-1779.; Salvi M., Vannucchi G., Campi I., Rossi S., Bonara P., Sbrozzi F., Guastella C., Avignone S., Pirola G., Ratiglia R., Beck-Peccoz P. Efficacy of rituximab treatment for thyroid-associated ophthalmopathy as a result of intraorbital B-cell depletion in one patient unresponsive to steroid immunosuppression // Eur. J. Endocrinol. – 2006. – Vol. 154, N 4. – P. 511-517.; Saigal K., Valencia I.C., Cohen J., Kerdel F.A. Hypocomplementemic urticarial vasculitis with angioedema, a rare presentation of systemic lupus erythematosus: rapid response to rituximab // J. Am. Acad. Dermatol. – 2003. – Vol. 49, N 5, Suppl. – P. S283-S285.; Levi M., Hack C.E., van Oers M.H. Rituximabinduced elimination of acquired angioedema due to C1-inhibitor deficiency // Am. J. Med. – 2006. – Vol. 119, N 8. – P. e3-e5.; Ratanatharathorn V., Ayash L., Reynolds C., Silver S., Reddy P., Becker M., Ferrara J.L., Uberti J.P. Treatment of chronic graft-versus-host disease with anti-CD20 chimeric monoclonal antibody // Biol. Blood Marrow Transplant. – 2003. – Vol. 9, N 8. – P. 505-511.; Okamoto M., Okano A., Akamatsu S., Ashihara E., Inaba T., Takenaka H., Katoh N., Kishimoto S., Shimazaki C. Rituximab is effective for steroid-refractory sclerodermatous chronic graftversus-host disease // Leukemia. – 2006. 20, N 1. – P. 172-173.; Cutler C., Miklos D., Kim H.T., Treister N., Woo S.B., Bienfang D., Klickstein L.B., Levin J., Miller K., Reynolds C., Macdonell R., Pasek M., Lee S.J., Ho V., Soiffer R., Antin J.H., Ritz J., Alyea E. Rituximab for steroid-refractory chronic graft-versus-host disease // Blood. – 2006. 108, N 2. – P. 756-762.; https://www.mimmun.ru/mimmun/article/view/116