New directions of pharmacotherapy of immune - inflammatory rheumatic diseases


Cite item

Full Text

Abstract

Deciphering immunopathogenesis, expanding the scope of diagnostics and developing new methods for treating human autoimmune diseases are among the priority areas of XXI century medicine. Particularly widely autoimmune pathology is presented in immunoinflammatory rheumatic diseases (IIRD), such as rheumatoid arthritis, systemic lupus erythematosus, systemic scleroderma, systemic vasculitis associated with the synthesis of antineutrophilic cytoplasmic antibodies, Sjogren's syndrome, idiopathic inflammatory myopathies and other other types of others. Deciphering the pathogenesis mechanisms of IIRD created the prerequisites for improving pharmacotherapy, which in the future should lead to a dramatic improvement in the prognosis for these diseases. The review discusses new approaches to IIRD pharmacotherapy associated with the inhibition of tumor necrosis factor-α, interleukin-6 (IL-6), IL-1β, IL-17, IL-23, and the prospects for using Janus kinase inhibitors, depending on the prevailing pathogenesis mechanisms - autoimmunity or autoinflammation.

About the authors

E L Nasonov

Nasonova Research Institute of Rheumatology; Sechenov First Moscow State Medical University (Sechenov University)

Email: nasonov@irramn.ru
д.м.н., академик РАН, научный руководитель ФГБНУ «НИИР им. В.А. Насоновой», проф. каф. ревматологии ИПО ФГАОУ ВО «Первый МГМУ им. И.М. Сеченова» Moscow, Russia

References

  1. Насонов Е.Л., Александрова Е.Н., Новиков А.А. Аутоиммунные ревматические заболевания - проблемы иммунопатологии и персонифицированной терапии. Вестник Российской академии медицинских наук. 2015;70(2):169-82.
  2. Wang L, Wang F.S, Gershwin M.E. Human autoimmune diseases: a comprehensive update. J Intern Med. 2015;278:369-95. doi: 10.1111/joim.12395
  3. Siebert S, Tsoukas A, Robertson J, Mc Innes I. Cytokines as therapeutic targets in rheumatoid arthritis and other inflammatory diseases. Pharmacol Rev. 2015;67(2):280-309. doi: 10.1124/pr.114.009639
  4. Baker K.F, Isaacs J.D. Novel therapies for immune - mediated inflammatory diseases: What can we learn from their use in rheumatoid arthritis, spondyloarthritis, systemic lupus erythematosus, psoriasis, Crohn's disease and ulcerative colitis? Ann Rheum Dis. 2018;77(2):175-87. doi: 10.1136/annrheumdis-2017-211555
  5. Насонов Е.Л. Фармакотерапия ревматоидного артрита: новая стратегия, новые мишени. Научно - практическая ревматология. 2017;55(4):409-19. doi: 10.14412/1995-4484-2017-409-419
  6. Rosenblum M.D, Remedios K.A, Abbas A.K. Mechanisms of human autoimmunity. J Clin Invest. 2015;125(6):2228-33. doi: 10.1172/JCI78088
  7. Theofilopoulos A.N, Kono D.H, Baccala R. The Multiple Pathways to Autoimmunity. Nat Immunol. 2017;18:716-24. doi: 10.1038/ni.3731
  8. Smolen J.S, Aletaha D, Mc Innes I.B. Rheumatoid arthritis. Lancet. 2016;22;388:2023-38. doi: 10.1016/S0140-6736(16)30173-8
  9. Mc-Innes I.B, Schett G. Pathogenetic insights from the treatment of rheumatoid arthritis. Lancet. 2017;389:2328-37.
  10. Насонов Е.Л. Проблемы иммунопатологии ревматоидного артрита: эволюция болезни. Научно - практическая ревматология. 2017;55(3):277-94. doi: 10.14412/1995-4484-2017-277-294
  11. Насонов Е.Л., ред. Генно - инженерные биологические препараты в лечении ревматоидного артрита. М.: ИМА-ПРЕСС, 2013.
  12. Smolen J.S, Aletaha D, Bijsma J.W.J, et al. For the T2T Expert Committee. Treating rheumatoid arthritis to target: recommendations of an international task force. Ann Rheum Dis. 2010;69:631-7. doi: 10.1136/ard.2009.123919
  13. Насонов Е.Л. Ревматоидный артрит. В кн.: Насонов Е.Л., ред. Российские клинические рекомендации. M.: ГЭОТАР-Медиа, 2017. C. 17-57.
  14. Monaco C, Nanchahal J, Taylor P, Feldmann M. Anti-TNF therapy: past, present and future. Int Immunol. 2015 Jan;27(1):55-62. doi: 10.1093/intimm/dxu102
  15. Kalliolias G.D, Ivashkiv L.B. TNF biology, pathogenetic mechanisms and emerging therapeutic strategies. Nat Rev Rheumatol. 2016;12:49-62.
  16. Boyman O, Comte D, Spertini F. Adverse reactions to biologic agents and their medical management. Nat Rev Rheumatol. 2014;10:612-27. doi: 10.1038/nrrheuum.2014.123
  17. Насонов Е.Л. Метотрексат при ревматоидном артрите - 2015: новые факты и идеи. Научно - практическая ревматология. 2015;53(4):421-33. doi: 10.14412/1995-4484-2015-421-433
  18. Hunter C.A, Jones S.A. IL-6 as a keystone cytokine in health and disease. Nat Immunol. 2015;15:448-57. doi: 10.1038/ni1117-1271b
  19. Garbers C, Heink S, Korn T, Rose-John S. Interleukin-6: designing specific therapeutics for a complex cytokine. Nat Rev Drug Discov. 2018;17(6):395-412. doi: 10.1038/nrd.2018.45
  20. Stone J.H, Tuckwell K, Dimonaco S, Klearman M, et al. Trial of Tocilizumab in Giant-Cell Arteritis. N Engl J Med. 2017;377(4):317-28. doi: 10.1056/NEJMoa1613849
  21. Сатыбалдыев А.М, Сатыбалдыева М.А, Насонов Е.Л. Тоцилизумаб в лечении ревматической полимиалгии и гигантоклеточного артериита. Клиническая фармакология и фармакотерапия. 2017;26(1):47-53.
  22. Бекетова Т.В, Насонов Е.Л. Инновационные методы лечения артериита Такаясу: в фокусе ингибиторы интерлейкина 6. Собственный опыт применения тоцилизумаба и обзор литературы. Научно - практическая ревматология. 2017;55(5):536-48. doi: 10.14412/1995-4484-2017-536-548
  23. Raimondo M.G, Biggioggero M, Crotti C, Becciolini A, Favalli E.G. Profile of sarilumab and its potential in the treatment of rheumatoid arthritis. Drug Des Devel Ther. 2017;11:1593-603. doi: 10.2147/DDDT.S100302
  24. June R.R, Olsen N.J. Room for more IL-6 blockade? Sarilumab for the treatment of rheumatoid arthritis. Expert Opin Biol Ther. 2016;16(10):1303-9. doi: 10.1080/14712598.2016.1217988
  25. Насонов Е.Л., Станислав М.Л., Раскина Т.А. и др. Эффективность и безопасность сарилумаба в комбинации с метотрексатом у пациентов с активным ревматоидным артритом и неадекватным эффектом монотерапии метотрексатом (результаты исследования III фазы по протоколу MOBILITY). Научно - практическая ревматология. 2019;57(2):142-8. doi: 10.14412/1995-4484-2019-142-148
  26. Emery P, van Hoogstraten H, Jayawardena S, Mangan EK, Cejas P, Verschueren P. Efficacy of Sarilumab in Patients with Rheumatoid Arthritis Who Previously Received Sarilumab or Tocilizumab [abstract]. Arthritis Rheum. 2017;69(suppl 10).
  27. Behrens E.M, Koretzky G.A. Review: Cytokine Storm Syndrome: Looking Toward the Precision Medicine Era. Arthritis Rheum. 2017;69(6):1135-43. doi: 10.1002/art.40071
  28. Насонов Е.Л., Лила А.М. Ингибиция интерлейкина 6 при иммуновоспалительных ревматических заболеваниях: достижения, перспективы и надежды. Научно - практическая ревматология. 2017;55(6):590-9. doi: 10.14412/1995-4484-2017-590-599
  29. Насонов Е.Л. Ингибиция иммунных контрольных точек и аутоиммунитет: ревматологические проблемы. Научно - практическая ревматология. 2018;56(1):5-9. doi: 10.14412/1995-4484-2018-5-9
  30. Wicks I.P, Roberts A.W. Targeting GM-CSF in inflammatory diseases. Nat Rev Rheumatol. 2016;12:37-48.
  31. Cook A.D, Hamilton J.A. Investigational therapies targeting the granulocyte macrophage colony - stimulating factor receptor - α in rheumatoid arthritis: focus on mavrilimumab. Ther Adv Musculoskelet Dis. 2018;10(2):29-38. doi: 10.1177/1759720X17752036
  32. Suurmond J, Diamond B. Autoantibodies in systemic autoimmune diseases: specificity and pathogenicity. J Clin Invest. 2015;125(6):2194-202. doi: 10.1172/JCI78084
  33. Насонов Е.Л., редактор. Анти-В-клеточная терапия в ревматологии: фокус на ритуксимаб. М.: ИМА-ПРЕСС, 2012. С. 119-52.
  34. Hofmann K, Clauder A.K, Manz R.A. Targeting B Cells and Plasma Cells in Autoimmune Diseases. Front Immunol. 2018 Apr 23;9:835.
  35. Schioppo T, Ingegnoli F. Current perspective on rituximab in rheumatic diseases. Drug Des Devel Ther. 2017;11:2891-904. doi: 10.2147/DDDT.S139248
  36. Насонов Е.Л. Перспективы анти-В-клеточной терапии в ревматологии. Научно - практическая ревматология. 2018;56(5):539-48. doi: 10.14412/1995-4484-2018-539-548
  37. Насонов Е.Л., Решетняк Т.М., Денисов Л.Н., Станислав М.Л., Соловьев С.К. Белимумаб: прогресс в лечении системной красной волчанки. Научно - практическая ревматология. 2012;50(5):13-9. doi: 10.14412/1995-4484-2012-1174
  38. Vincent F.B, Morand E.F, Schneider P, Mackay F. The BAFF/APRIL system in SLE pathogenesis. Nat Rev Rheumatol. 2014;10(6):365-73.
  39. Sanz I, Yasothan U, Kirkpatrick P. Belimumab. Nat Rev Drug Discov. 2011;10(5):335-6.
  40. Асеева Е.А., Соловьев С.К., Меснянкина А.А., Решетняк Т.М., Лопатина Н.Е., Глухова С.И., Насонов Е.Л. Результаты наблюдательного проспективного исследования эффективности и безопасности белимумаба (Бенлисты®) при системной красной волчанке в реальной клинической практике. Научно - практическая ревматология. 2016;54(1):31-7. doi: 10.14412/1995-4484-2016-31-37
  41. Ehrenstein M.R, Wing C. The BAFFling effects of rituximab in lupus: danger ahead? Nat Rev Rheumatol. 2016;12:367-72.
  42. Меснянкина А.А., Соловьев С.К., Александрова Е.Н., Алексанкин А.П., Асеева Е.А., Насонов Е.Л. Двойная терапия генно - инженерными биологическими препаратами у пациентов с системной красной волчанкой. Научно - практическая ревматология. 2016;54(3):281-8. doi: 10.14412/1995-4484-2016-281-288
  43. Меснянкина А.А., Соловьев С.К., Асеева Е.А., Насонов Е.Л. Эффективность генно - инженерной биологической терапии и особенности гуморального иммунитета у больных системной красной волчанкой. Научно - практическая ревматология. 2018;56(3):302-9. doi: 10.14412/1995-4484-2018-302-309
  44. Tak P.P, Thurlings R.M, Rossier C, Nestorov I, Dimic A, Mircetic V, Rischmueller M, Nasonov E, Shmidt E, Emery P, Munafo A. Atacicept in patients with rheumatoid arthritis: results of a multicenter, phase Ib, double - blind, placebo - controlled, dose - escalating, single - and repeated - dose study. Arthritis Rheum. 2008;58(1):61-72.
  45. Pena-Rossi C, Nasonov E, Stanislav M, Yakusevich V, Ershova O, Lomareva N, Saunders H, Hill J, Nestorov I. An exploratory dose - escalating study investigating the safety, tolerability, pharmacokinetics and pharmacodynamics of intravenous atacicept in patients with systemic lupus erythematosus. Lupus. 2009;18(6):547-55.
  46. Merrill J.T, Wallace D.J, Wax S, Kao A, Fraser P.A, et al.; ADDRESS II Investigators. Efficacy and Safety of Atacicept in Patients With Systemic Lupus Erythematosus: Results of a Twenty-Four-Week, Multicenter, Randomized, Double-Blind, Placebo-Controlled, Parallel-Arm, Phase IIb Study. Arthritis Rheum. 2018;70(2):266-76.
  47. Hiepe F, Dörner T, Hauser A.E, Hoyer B.F, Mei H, Radbruch A. Long - lived autoreactive plasma cells drive persistent autoimmune inflammation. Nat Rev Rheumatol. 2011;7(3):170-8. doi: 10.1038/nrrheum.2011.1
  48. Verbrugge S.E, Scheper R.J, Lems W.F, de Gruijl T.D, Jansen G. Proteasome inhibitors as experimental therapeutics of autoimmune diseases. Arthritis Res Ther. 2015;17:17. doi: 10.1186/s13075-015-0529-1
  49. Mc Gonagle D, Mc Dermott M.F. A Proposed Classification of the Immunological Diseases. PLoS Med. 2006;3:e297. doi: 10.1371/journal.pmed.0030297
  50. Banchereau R, Cepika A.M, Banchereau J, Pascual V. Understanding Human Autoimmunity and Autoinflammation Through Transcriptomics. Annu Rev Immunol. 2017;35:337-70. doi: 10.1146/annurev-immunol-051116-052225
  51. Dinarello C.A. An expanding role for interleukin-1 blockade from gout to cancer. Mol Med. 2014;20 (Suppl 1):S43-S58.
  52. Lopalco G, Cantarini L, Vitale A, et al. Interleukin-1 as common denominator from autoinflammatory to autoimmune disorders: premises, perils, and perspectives. Mediat Inflamm. 2015;2015:194864.
  53. Насонов Е.Л., Елисеев М.С. Роль интерлейкина 1 в развитии заболеваний человека. Научно - практическая ревматология. 2016;54(1):60-77. doi: 10.14412/1995-4484-2016-60-77
  54. Schett G, Dayer J-M, Manger B. Interleukin-1 function and role in rheumatic disease. Nat Rev Rheumatol. 2016 Jan;12(1):14-24. doi: 10.1038/nrrheum.2016.166
  55. Mantovani A, Dinarello C.A, Molgora M, Garlanda C. Interleukin-1 and Related Cytokines in the Regulation of Inflammation and Immunity. Immunity. 2019 Apr 16;50(4):778-95. doi: 10.1016/j.immuni.2019.03.012
  56. Guo H, Callaway J.B, Jenny P-Y, Ting J P-Y. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med. 2015;21:677-87. doi: 10.1038/nm.3893
  57. Елисеев М.С., Насонов Е.Л. Применение канакинумаба при подагре. Научно - практическая ревматология. 2018;56(Прил. 4):41-8. doi: 10.14412/1995-4484-2018-41-48
  58. Bettiol A, Lopalco G, Emmi G, Cantarini L, Urban M.L, et al. Unveiling the Efficacy, Safety, and Tolerability of Anti - Interleukin-1 Treatment in Monogenic and Multifactorial Autoinflammatory Diseases. Int J Mol Sci. 2019 Apr 17;20(8). pii: E1898. doi: 10.3390/ijms20081898
  59. Hausmann J.S. Targeting cytokines to treat autoinflammatory diseases. Clin Immunol. 2018 Oct 28. pii: S1521-6616(18)30432-7. doi: 10.1016/j.clim.2018.10.016
  60. Libby P. Interleukin-1 Beta as a Target for Atherosclerosis Therapy: Biological Basis of CANTOS and Beyond. J Am Coll Cardiol. 2017;70(18):2278-89. doi: 10.1016/j.jacc.2017.09.028
  61. Ridker P.M, Everett B.M, Thuren T, et al; CANTOS Trial Group. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N Engl J Med. 2017;377(12):1119-31. doi: 10.1056/NEJMoa1707914
  62. Ridker P.M, Mac Fadyen J.G, Everett B.M, Libby P, Thuren T, Glynn R.J; CANTOS Trial Group. Relationship of C-reactive protein reduction to cardiovascular event reduction following treatment with canakinumab: a secondary analysis from the CANTOS randomised controlled trial. Lancet. 2018;391(10118):319-28. doi: 10.1016/S0140-6736(17)32814-3
  63. Fragoulis G.E, Siebert S, McInnes I.B. Therapeutic targeting of IL-17 and IL-23 cytokines in immune - mediated disease. Ann Rev Med. 2016;67:337-53. doi: 10.1146/annurev-med-051914-0219444
  64. Lubberts E. The IL-23-IL-17 axis in inflammatory arthritis. Nat Rev Rheumatol. 2015;11(7):415-29. doi: 10.1038/nrrheum.2015.53
  65. Bianchi E, Rogge L. The IL-23/IL-17 pathway in human chronic inflammatory diseases - new insight from genetics and targeted therapies. Genes Immun. 2019;20(5):415-25. doi: 10.1038/s41435-019-0067-y
  66. Mc Innes I.B, Porter D, Siebert S. Choosing new targets for rheumatoid arthritis therapeutics: too interesting to Fail? Arthritis Rheum. 2017;69:1131-4.
  67. Blauvelt A, Chiricozzi A. The Immunologic Role of IL-17 in Psoriasis and Psoriatic Arthritis Pathogenesis. Clin Rev Allergy Immunol. 2018 Dec;55(3):379-90. doi: 10.1007/s12016-018-8702-3
  68. Mease P.J. Inhibition of interleukin-17, interleukin-23 and the TH17 cell pathway in the treatment of psoriatic arthritis and psoriasis. Curr Opin Rheumatol. 2015 Mar;27(2):127-33. doi: 10.1097/BOR.000
  69. Насонов Е.Л. Новые возможности фармакотерапии иммуновоспалительных ревматических заболеваний: фокус на ингибиторы интерлейкина 17. Научно - практическая ревматология. 2017;55(1):68-86. doi: 10.14412/1995-4484-2017-68-86
  70. Pfeifle R, Rothe T, Ipseiz N, et al. Regulation of autoantibody activity by the IL-23-TH17 axis determines the onset of autoimmune disease. Nat Immunol. 2017;18:104-13. doi: 10.1038/ni.3579
  71. Schwartz D.M, Kanno Y, Villarino A, Ward M, Gadina M, O'Shea J.J. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat Rev Drug Discov. 2017;16(12):843-62. doi: 10.1038/nrd.2017.201
  72. Gadina M, Johnson C, Schwartz D, Bonelli M, Hasni S, Kanno Y, Changelian P, Laurence A, O'Shea J.J. Translational and clinical advances in JAK-STAT biology: The present and future of jakinibs. J Leukoc Biol. 2018;104(3):499-514. doi: 10.1002/JLB.5RI0218-084R
  73. Насонов ЕЛ. Новые подходы к фармакотерапии ревматоидного артрита: тофацитиниб. Научно - практическая ревматология. 2014;52(2):209-21. doi: 10.14412/1995-4484-2014-209-221
  74. Dhillon S. Tofacitinib: A Review in Rheumatoid Arthritis. Drugs. 2017 Dec;77(18):1987-2001. doi: 10.1007/s40265-017-0835-9
  75. Berekmeri A, Mahmood F, Wittmann M, Helliwell P. Tofacitinib for the treatment of psoriasis and psoriatic arthritis. Expert Rev Clin Immunol. 2018;14(9):719-30. doi: 10.1080/1744666X.2018
  76. Veale D.J, Mc Gonagle D, Mc Innes I.B, Krueger J.G, Ritchlin C.T, Elewaut D, Kanik K.S, Hendrikx T, Berstein G, Hodge J, Telliez J.B. The rationale for Janus kinase inhibitors for the treatment of spondyloarthritis. Rheumatology (Oxford). 2018 Apr 3. doi: 10.1093/rheumatology/key070
  77. Cinats A, Heck E, Robertson L. Janus Kinase Inhibitors: A Review of Their Emerging Applications in Dermatology. Skin Ther Lett. 2018y;23(3):5-9.
  78. Soendergaard C, Bergenheim F.H, Bjerrum J.T, Nielsen O.H. Targeting JAK-STAT signal transduction in IBD. Pharmacol Ther. 2018 Jul 23. pii: S0163-7258(18)30122-0. doi: 10.1016/j.pharmthera.2018.07.003
  79. Насонов Е.Л, Абдулганиева Д.И, Файрушина И.Ф. Место тофацитиниба в лечении воспалительных заболеваний кишечника. Терапевтический архив. 2019;91(2):101-8. doi: 10.26442/00403660.2019.02.000155
  80. Hoffman H.M, Broderick L. JAK inhibitors in autoinflammation. J Clin Invest. 2018 Jul 2;128(7):2760-2. doi: 10.1172/JCI121526
  81. Mok C.C. The Jakinibs in systemic lupus erythematosus: progress and prospects. Expert Opin Investig Drugs. 2019;28(1):85-92. doi: 10.1080/13543784.2019.1551358
  82. Kahn J.S, Deverapalli S.C, Rosmarin D.M. JAK-STAT signaling pathway inhibition: a role for treatment of discoid lupus erythematosus and dermatomyositis. Int J Dermatol. 2018;57(8):1007-14. doi: 10.1111/ijd.14064
  83. Ruperto N, Brunner H.I, Zuber Z, Tzaribachev N, Kingsbury D.J, Foeldvari I, et al. Pharmacokinetic and safety profile of tofacitinib in children with polyarticular course juvenile idiopathic arthritis: results of a phase 1, open - label, multicenter study. Pediatr Rheumatol. 2017;15:86. doi: 10.1186/s12969-017-0212-y
  84. Насонов Е.Л., Лила А.М. Ингибиторы Янус - киназ при иммуновоспалительных ревматических заболеваниях: новые возможности и перспективы. Научно - практическая ревматология. 2019;57(1):8-16. doi: 10.14412/1995-4484-2019-8-16
  85. Virtanen A, Haikarainen T, Raivola J, Silvennoinen O. Selective JAKinibs: Prospects in Inflammatory and Autoimmune Diseases. BioDrugs. 2019;33(1):15-32. doi: 10.1007/s40259-019-00333-w
  86. Smolen J.S, van der Heijde D, Machold K.P, Aletaha D, Landewé R. Proposal for a new nomenclature of disease - modifying antirheumatic drugs. Ann Rheum Dis. 2014;73(1):3-5. doi: 10.1136/annrheumdis-2013-204317
  87. Winthrop K.L. The emerging safety profile of JAK inhibitors in rheumatic disease. Nat Rev Rheumatol. 2017;13(4):234-43. doi: 10.1038/nrrheum.2017.23
  88. Nash P, Coates L.C, Fleischmann R, Papp K.A, Gomez-Reino J.J, Kanik K.S, Wang C, Wu J, Menon S, Hendrikx T, Ports W.C. Efficacy of Tofacitinib for the Treatment of Psoriatic Arthritis: Pooled Analysis of Two Phase 3 Studies. Rheumatol Ther. 2018;5(2):567-82. doi: 10.1007/s40744-018-0131-5
  89. Tian F, Chen Z, Xu T. Efficacy and safety of tofacitinib for the treatment of chronic plaque psoriasis: a systematic review and meta - analysis. J Int Med Res. 2019 May 17:300060519847414. doi: 10.1177/0300060519847414
  90. Singh J.A, Guyatt G, Ogdie A, Gladman D.D, Deal C, et al. Special Article: 2018 American College of Rheumatology/National Psoriasis Foundation Guideline for the Treatment of Psoriatic Arthritis. Arthritis Rheum. 2019;71(1):5-32. doi: 10.1002/art.40726
  91. Van der Heijde D, Deodhar A, Wei J.C, et al. Tofacitinib in patients with ankylosing spondylitis: a phase II, 16-week, randomised, placebo - controlled, dose - ranging study. Ann Rheum Dis. 2017;76:1340-7.
  92. Maksymowych W.P, Heijde D.V, Baraliakos X, Deodhar A, Sherlock S.P, Li D, Fleishaker D, Hendrikx T, Kanik K.S. Tofacitinib is associated with attainment of the minimally important reduction in axial magnetic resonance imaging inflammation in ankylosing spondylitis patients. Rheumatology (Oxford). 2018;57(8):1390-9. doi: 10.1093/rheumatology/key104
  93. Kunwar S, Collins C.E, Constantinescu F. Baricitinib, a Janus kinase inhibitor, in the treatment of rheumatoid arthritis: a systematic literature review and meta - analysis of randomized controlled trials. Clin Rheumatol. 2018 Jul 13. doi: 10.1007/s10067-018-4199-7
  94. Al-Salama Z.T, Scott L.J. Baricitinib: A Review in Rheumatoid Arthritis. Drugs. 2018;78(7):761-72. doi: 10.1007/s40265-018-0908-4
  95. Taylor P.C, Keystone E.C, van der Heijde D, Weinblatt M.E, Del Carmen Morales L, et al. Baricitinib versus Placebo or Adalimumab in Rheumatoid Arthritis. N Engl J Med. 2017;376(7):652-62. doi: 10.1056/NEJMoa1608345
  96. Takeuchi T, Genovese M.C, Haraoui B, Li Z, Xie L, et al. Dose reduction of baricitinib in patients with rheumatoid arthritis achieving sustained disease control: results of a prospective study. Ann Rheum Dis. 2019;78(2):171-8. doi: 10.1136/annrheumdis-2018-213271
  97. Damsky W, King B.A. JAK inhibitors in dermatology: The promise of a new drug class. J Am Acad Dermatol. 2017;76(4):736-44. doi: 10.1016/j.jaad.2016.12.005
  98. Hosking A.M, Juhasz M, Mesinkovska N.A. Topical Janus kinase inhibitors: A review of applications in dermatology. J Am Acad Dermatol. 2018;79(3):535-44. doi: 10.1016/j.jaad.2018.04.018
  99. Kretschmer S, Lee-Kirsch M.A. Type I interferon - mediated autoinflammation and autoimmunity. Curr Opin Immunol. 2017;49:96-102. doi: 10.1016/j.coi.2017.09.003
  100. Sanchez G.A.M, Reinhardt A, Ramsey S, Wittkowski H, Hashkes P.J, et al. JAK1/2 inhibition with baricitinib in the treatment of autoinflammatory interferonopathies. J Clin Invest. 2018;128(7):3041-52. doi: 10.1172/JCI98814
  101. König N, Fiehn C, Wolf C, Schuster M, Cura Costa E, Tüngler V, et al. Familial chilblain lupus due to a gain - of - function mutation in STING. Ann Rheum Dis. 2017;76(2):468-72.
  102. Rodero M.P, Frémond M-L, Rice G.I, Neven B, Crow Y.J. JAK inhibition in STING-associated interferonopathy. Ann Rheum Dis. 2016;75(12):e75.
  103. Seo J, Kang J-A, Dong In Suh D, et al. Tofacitinib relieves symptoms of stimulator of interferon genes (STING)-associated vasculopathy with onset in infancy caused by 2 de novo variants in TMEM173. J Allergy Clin Immunol. 2017;139(4):1396-9.e12. doi: 10.1016/j.jaci.2016.10.030
  104. Psarras A, Emery P, Vital E.M. Type I interferon - mediated autoimmune diseases: pathogenesis, diagnosis and targeted therapy. Rheumatology (Oxford). 2017;56(10):1662-75. doi: 10.1093/rheumatology/kew431
  105. Choy E.H. Clinical significance of Janus Kinase inhibitor selectivity. Rheumatology (Oxford). 2018 Dec 1. doi: 10.1093/rheumatology/key339
  106. Boutet M.A, Nerviani A, Gallo Afflitto G, Pitzalis C. Role of the IL-23/IL-17 Axis in Psoriasis and Psoriatic Arthritis: The Clinical Importance of Its Divergence in Skin and Joints. Int J Mol Sci. 2018;19(2). pii: E530. doi: 10.3390/ijms19020530
  107. Gaston J.S.H, Jadon D.R. Th17 cell responses in spondyloarthritis. Best Pract Res Clin Rheumatol. 2017;31(6):777-96. doi: 10.1016/j.berh.2018.07.010
  108. Verstockt B, van Assche G, Vermeire S, Ferrante M. Biological therapy targeting the IL-23/IL-17 axis in inflammatory bowel disease. Expert Opin Biol Ther. 2017;17(1):31-47.
  109. Papp K, Gordon K, Thaçi D, Morita A, et al. Phase 2 trial of selective tyrosine kinase 2 inhibition in psoriasis. N Engl J Med. 2018;379(14):1313-21. doi: 10.1056/NEJMoa1806382
  110. Deng G.M, Kyttaris V.C, Tsokos G.C. Targeting Syk in Autoimmune Rheumatic Diseases. Front Immunol. 2016;7:78. doi: 10.3389/fimmu.2016.00

Copyright (c) 2019 Consilium Medicum

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License.
 
 


This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies