Skip to main content
Top
Published in: Current Rheumatology Reports 12/2018

Open Access 01-12-2018 | Pediatric Rheumatology (S Ozen, Section Editor)

JAK Inhibitors in Rheumatology: Implications for Paediatric Syndromes?

Authors: S. A. Kerrigan, I. B. McInnes

Published in: Current Rheumatology Reports | Issue 12/2018

Login to get access

Abstract

Purpose of Review

Given the recent increase in the profile and use of Janus kinase inhibitors (JAKinibs) in adult patients with rheumatic diseases, we aimed to review the current evidence accruing for use in paediatric rheumatology patients.

Recent Findings

Significant advances have been made in the management of rheumatic diseases in the past two decades. The introduction of biologic agents in both adults and children has provided significant improvements to patient outcomes and led to better quality of life. Moreover, responses to similar agents allude to common effector pathways operating across juvenile and adult synovitis especially. However, inefficacy and intolerance of these agents leads to a subset of children with limited treatment options.

Summary

Since 2012, Janus kinase (JAK) inhibitors (JAKinibs), a novel group of oral small molecule inhibitors, have demonstrated their efficacy in several forms of adult inflammatory arthritis, such as rheumatoid arthritis (RA) and psoriatic arthritis (PsA). There are hopes that these successes will be transferable to the paediatric population. In the following review, we discuss the development and progress of JAKinibs in this regard.
Literature
1.
go back to reference Schwartz DM, Bonelli M, Gadina M, O'Shea JJ. Type I/II cytokines, JAKs, and new strategies for treating autoimmune diseases. Nature Publishing Group; 2015;12(1):25–36. Schwartz DM, Bonelli M, Gadina M, O'Shea JJ. Type I/II cytokines, JAKs, and new strategies for treating autoimmune diseases. Nature Publishing Group; 2015;12(1):25–36.
2.
go back to reference Schwartz DM, Kanno Y, Villarino A, Ward M, Gadina M, O'Shea JJ. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nature Publishing Group; 2017;17(1):843–62. Schwartz DM, Kanno Y, Villarino A, Ward M, Gadina M, O'Shea JJ. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nature Publishing Group; 2017;17(1):843–62.
3.
go back to reference Fleischmann R, Mysler E, Hall S, Kivitz AJ, Moots RJ, Luo Z, et al. Efficacy and safety of tofacitinib monotherapy, tofacitinib with methotrexate, and adalimumab with methotrexate in patients with rheumatoid arthritis (ORAL Strategy): a phase 3b/4, double-blind, head-to-head, randomised controlled trial. Lancet. Elsevier Ltd; 2017;390(10093):457–68. Fleischmann R, Mysler E, Hall S, Kivitz AJ, Moots RJ, Luo Z, et al. Efficacy and safety of tofacitinib monotherapy, tofacitinib with methotrexate, and adalimumab with methotrexate in patients with rheumatoid arthritis (ORAL Strategy): a phase 3b/4, double-blind, head-to-head, randomised controlled trial. Lancet. Elsevier Ltd; 2017;390(10093):457–68.
4.
go back to reference Fleischmann R, Kremer J, Cush J, Schulze-Koops H, Connell CA, Bradley JD, et al. Placebo-controlled trial of tofacitinib monotherapy in rheumatoid arthritis. N Engl J Med. 2012;367(6):495–507.CrossRef Fleischmann R, Kremer J, Cush J, Schulze-Koops H, Connell CA, Bradley JD, et al. Placebo-controlled trial of tofacitinib monotherapy in rheumatoid arthritis. N Engl J Med. 2012;367(6):495–507.CrossRef
5.
go back to reference Vieira M-C, Zwillich SH, Jansen JP, Smiechowski B, Spurden D, Wallenstein GV. Tofacitinib versus biologic treatments in patients with active rheumatoid arthritis who have had an inadequate response to tumor necrosis factor inhibitors_ results from a network meta-analysis. Clin Ther. Elsevier HS Journals, Inc; 2016;38(12):2628–2641.e5. Vieira M-C, Zwillich SH, Jansen JP, Smiechowski B, Spurden D, Wallenstein GV. Tofacitinib versus biologic treatments in patients with active rheumatoid arthritis who have had an inadequate response to tumor necrosis factor inhibitors_ results from a network meta-analysis. Clin Ther. Elsevier HS Journals, Inc; 2016;38(12):2628–2641.e5.
6.
go back to reference Eisenstein EM, Berkun Y. Diagnosis and classification of juvenile idiopathic arthritis. J Autoimmun. Elsevier Ltd; 2014;48-49(c):31–3. Eisenstein EM, Berkun Y. Diagnosis and classification of juvenile idiopathic arthritis. J Autoimmun. Elsevier Ltd; 2014;48-49(c):31–3.
7.
go back to reference Giancane G, Consolaro A, Lanni S, Davì S, Schiappapietra B, Ravelli A. Juvenile idiopathic arthritis: diagnosis and treatment. Rheumatol Ther. 2016;3(2):187–207. Giancane G, Consolaro A, Lanni S, Davì S, Schiappapietra B, Ravelli A. Juvenile idiopathic arthritis: diagnosis and treatment. Rheumatol Ther. 2016;3(2):187–207.
8.
go back to reference Clark JD, Flanagan ME, Telliez J-B. Discovery and development of Janus kinase (JAK) inhibitors for inflammatory diseases. J Med Chem. 2014;57(12):5023–38.CrossRef Clark JD, Flanagan ME, Telliez J-B. Discovery and development of Janus kinase (JAK) inhibitors for inflammatory diseases. J Med Chem. 2014;57(12):5023–38.CrossRef
9.
go back to reference Banerjee S, Biehl A, Gadina M, Hasni S, Schwartz DM. JAK–STAT signaling as a target for inflammatory and autoimmune diseases: current and future prospects. Drugs. Springer International Publishing; 2017;77(5):521–46. Banerjee S, Biehl A, Gadina M, Hasni S, Schwartz DM. JAK–STAT signaling as a target for inflammatory and autoimmune diseases: current and future prospects. Drugs. Springer International Publishing; 2017;77(5):521–46.
10.
go back to reference Kudlacz E, Perry B, Sawyer P, Conklyn M, McCurdy S, Brissette W, et al. The novel JAK-3 inhibitor CP-690550 is a potent immunosuppressive agent in various murine models. Am J Transplant. Wiley/Blackwell (10.1111); 2004;4(1):51–7. Kudlacz E, Perry B, Sawyer P, Conklyn M, McCurdy S, Brissette W, et al. The novel JAK-3 inhibitor CP-690550 is a potent immunosuppressive agent in various murine models. Am J Transplant. Wiley/Blackwell (10.1111); 2004;4(1):51–7.
11.
go back to reference • Prahalad S, Conneely KN, Jiang Y, Sudman M, Wallace CA, Brown MR, et al. Brief report: susceptibility to childhood-onset rheumatoid arthritis: investigation of a weighted genetic risk score that integrates cumulative effects of variants at five genetic loci. Arthritis Rheum. 2013;65(6):1663–7 This study nicely illustrates the genetic similarities between rheumatoid arthritis and RF+ve JIA.CrossRef • Prahalad S, Conneely KN, Jiang Y, Sudman M, Wallace CA, Brown MR, et al. Brief report: susceptibility to childhood-onset rheumatoid arthritis: investigation of a weighted genetic risk score that integrates cumulative effects of variants at five genetic loci. Arthritis Rheum. 2013;65(6):1663–7 This study nicely illustrates the genetic similarities between rheumatoid arthritis and RF+ve JIA.CrossRef
12.
go back to reference Barnes MG, Grom AA, Thompson SD, Griffin TA, Pavlidis P, Itert L, et al. Subtype-specific peripheral blood gene expression profiles in recent-onset juvenile idiopathic arthritis. Arthritis Rheum. 2009;60(7):2102–12.CrossRef Barnes MG, Grom AA, Thompson SD, Griffin TA, Pavlidis P, Itert L, et al. Subtype-specific peripheral blood gene expression profiles in recent-onset juvenile idiopathic arthritis. Arthritis Rheum. 2009;60(7):2102–12.CrossRef
13.
go back to reference Ravelli A, Felici E, Magni-Manzoni S, Pistorio A, Novarini C, Bozzola E, et al. Patients with antinuclear antibody-positive juvenile idiopathic arthritis constitute a homogeneous subgroup irrespective of the course of joint disease. Arthritis Rheum. Wiley-Blackwell; 2005;52(3):826–32. Ravelli A, Felici E, Magni-Manzoni S, Pistorio A, Novarini C, Bozzola E, et al. Patients with antinuclear antibody-positive juvenile idiopathic arthritis constitute a homogeneous subgroup irrespective of the course of joint disease. Arthritis Rheum. Wiley-Blackwell; 2005;52(3):826–32.
14.
go back to reference Mellins ED, Macaubas C, Grom AA. Pathogenesis of systemic juvenile idiopathic arthritis: some answers, more questions. Nature Publishing Group; 2011;7(7):416–26. Mellins ED, Macaubas C, Grom AA. Pathogenesis of systemic juvenile idiopathic arthritis: some answers, more questions. Nature Publishing Group; 2011;7(7):416–26.
15.
go back to reference Grom AA, Horne A, De Benedetti F. Macrophage activation syndrome in the era of biologic therapy. Nature Publishing Group; 2016;12(5):259–68. Grom AA, Horne A, De Benedetti F. Macrophage activation syndrome in the era of biologic therapy. Nature Publishing Group; 2016;12(5):259–68.
16.
go back to reference O'Neill LAJ, Golenbock D, Bowie AG. The history of Toll-like receptors — redefining innate immunity. Nature Publishing Group; 2013;13(6):453–60. O'Neill LAJ, Golenbock D, Bowie AG. The history of Toll-like receptors — redefining innate immunity. Nature Publishing Group; 2013;13(6):453–60.
17.
go back to reference Bonjardim CA, Ferreira PCP, Kroon EG. Interferons: signaling, antiviral and viral evasion. Immunol Lett. 2009;122(1):1–11.CrossRef Bonjardim CA, Ferreira PCP, Kroon EG. Interferons: signaling, antiviral and viral evasion. Immunol Lett. 2009;122(1):1–11.CrossRef
18.
go back to reference •• Volpi S, Picco P, Caorsi R, Candotti F, Gattorno M. Type I interferonopathies in pediatric rheumatology. Pediatr Rheumatol; 2017; 1–12. This review gives a detailed background description of interferon signaling and interferonopathies. •• Volpi S, Picco P, Caorsi R, Candotti F, Gattorno M. Type I interferonopathies in pediatric rheumatology. Pediatr Rheumatol; 2017; 1–12. This review gives a detailed background description of interferon signaling and interferonopathies.
19.
go back to reference Sun L, Wu J, Du F, Chen X, Chen ZJ. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science. 2013;339(6121):786–91.CrossRef Sun L, Wu J, Du F, Chen X, Chen ZJ. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science. 2013;339(6121):786–91.CrossRef
20.
go back to reference Crow YJ. Type I interferonopathies: a novel set of inborn errors of immunity. Annals of the New York Academy of Sciences. 2017. pp. 1–8. Crow YJ. Type I interferonopathies: a novel set of inborn errors of immunity. Annals of the New York Academy of Sciences. 2017. pp. 1–8.
21.
go back to reference Tolmie JL, Shillito P, Hughes-Benzie R, Stephenson JBP. The Aicardi-Goutières syndrome (familial, early onset encephalopathy with calcifications of the basal ganglia and chronic cerebrospinal fluid lymphocytosis). J Med Genet. 2006;:1–4. Tolmie JL, Shillito P, Hughes-Benzie R, Stephenson JBP. The Aicardi-Goutières syndrome (familial, early onset encephalopathy with calcifications of the basal ganglia and chronic cerebrospinal fluid lymphocytosis). J Med Genet. 2006;:1–4.
22.
go back to reference • Livingston J, Crow Y. Neurologic phenotypes associated with mutations in TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, ADAR1, and IFIH1: Aicardi–Goutières Syndrome and Beyond. Neuropediatrics. 2016;47(06):355–60 A thorough descriptor of genotypic and corresponding phenotypic subtypes of AGS.CrossRef • Livingston J, Crow Y. Neurologic phenotypes associated with mutations in TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, ADAR1, and IFIH1: Aicardi–Goutières Syndrome and Beyond. Neuropediatrics. 2016;47(06):355–60 A thorough descriptor of genotypic and corresponding phenotypic subtypes of AGS.CrossRef
23.
go back to reference Richards A, van den Maagdenberg AMJM, Jen JC, Kavanagh D, Bertram P, Spitzer D, et al. C-terminal truncations in human 3′-5′ DNA exonuclease TREX1 cause autosomal dominant retinal vasculopathy with cerebral leukodystrophy. Nat Gen. 2007;39(9):1068–70.CrossRef Richards A, van den Maagdenberg AMJM, Jen JC, Kavanagh D, Bertram P, Spitzer D, et al. C-terminal truncations in human 3′-5′ DNA exonuclease TREX1 cause autosomal dominant retinal vasculopathy with cerebral leukodystrophy. Nat Gen. 2007;39(9):1068–70.CrossRef
24.
go back to reference Navarro V, Scott C, Briggs TA, Barete S, Frances C, Lebon P, et al. Two further cases of spondyloenchondrodysplasia (SPENCD) with immune dysregulation. Am J Med Genet A. 2008;146A(21):2810–5.CrossRef Navarro V, Scott C, Briggs TA, Barete S, Frances C, Lebon P, et al. Two further cases of spondyloenchondrodysplasia (SPENCD) with immune dysregulation. Am J Med Genet A. 2008;146A(21):2810–5.CrossRef
25.
go back to reference Liu Y, Jesus AA, Marrero B, Yang D, Ramsey SE, Montealegre Sanchez GA, et al. Activated STING in a Vascular and Pulmonary Syndrome. N Engl J Med. 2014;371(6):507–18.CrossRef Liu Y, Jesus AA, Marrero B, Yang D, Ramsey SE, Montealegre Sanchez GA, et al. Activated STING in a Vascular and Pulmonary Syndrome. N Engl J Med. 2014;371(6):507–18.CrossRef
26.
go back to reference Meuwissen MEC, Schot R, Buta S, Oudesluijs G, Tinschert S, Speer SD, et al. Human USP18 deficiency underlies type 1 interferonopathy leading to severe pseudo-TORCH syndrome. J Exp Med. Rockefeller University Press; 2016;213(7):1163–74. Meuwissen MEC, Schot R, Buta S, Oudesluijs G, Tinschert S, Speer SD, et al. Human USP18 deficiency underlies type 1 interferonopathy leading to severe pseudo-TORCH syndrome. J Exp Med. Rockefeller University Press; 2016;213(7):1163–74.
27.
go back to reference Singleton EB, Merten DF. An unusual syndrome of widened medullary cavities of the metacarpals and phalanges, aortic calcification and abnormal dentition. Pediatr Radiol. 1973;1(1):2–7.CrossRef Singleton EB, Merten DF. An unusual syndrome of widened medullary cavities of the metacarpals and phalanges, aortic calcification and abnormal dentition. Pediatr Radiol. 1973;1(1):2–7.CrossRef
28.
go back to reference Rutsch F, MacDougall M, Lu C, Buers I, Mamaeva O, Nitschke Y, et al. A Specific IFIH1 Gain-of-function mutation causes Singleton-Merten Syndrome. Am J Hum Genet. 2015;96(2):275–82.CrossRef Rutsch F, MacDougall M, Lu C, Buers I, Mamaeva O, Nitschke Y, et al. A Specific IFIH1 Gain-of-function mutation causes Singleton-Merten Syndrome. Am J Hum Genet. 2015;96(2):275–82.CrossRef
29.
go back to reference Torrelo A CANDLE syndrome as a paradigm of proteasome-related autoinflammation. Front Immunol 2017;8:489–9. Torrelo A CANDLE syndrome as a paradigm of proteasome-related autoinflammation. Front Immunol 2017;8:489–9.
30.
go back to reference Lee WS, Teo KM, Ng RT, Chong SY, Kee BP, Chua KH. Novel mutations in SKIV2L and TTC37 genes in Malaysian children with trichohepatoenteric syndrome. Gene. 2016;586(1):1–6.CrossRef Lee WS, Teo KM, Ng RT, Chong SY, Kee BP, Chua KH. Novel mutations in SKIV2L and TTC37 genes in Malaysian children with trichohepatoenteric syndrome. Gene. 2016;586(1):1–6.CrossRef
31.
go back to reference Pezzani L, Brena M, Callea M, Colombi M, Tadini G. X-linked reticulate pigmentary disorder with systemic manifestations: a new family and review of the literature. Am J Med Genet A. Wiley-Blackwell; 2013;161A(6):1414–20. Pezzani L, Brena M, Callea M, Colombi M, Tadini G. X-linked reticulate pigmentary disorder with systemic manifestations: a new family and review of the literature. Am J Med Genet A. Wiley-Blackwell; 2013;161A(6):1414–20.
32.
go back to reference Davidson S, Steiner A, Harapas CR, Masters SL. An update on autoinflammatory diseases: interferonopathies. Curr Rheumatol Rep. Springer US; 2018;20(7):38. Davidson S, Steiner A, Harapas CR, Masters SL. An update on autoinflammatory diseases: interferonopathies. Curr Rheumatol Rep. Springer US; 2018;20(7):38.
33.
go back to reference Fiehn C Familial chilblain lupus - what can we learn from type I interferonopathies? Curr Rheumatol Rep; 2017;:1–6. Fiehn C Familial chilblain lupus - what can we learn from type I interferonopathies? Curr Rheumatol Rep; 2017;:1–6.
34.
go back to reference Lee-Kirsch MA, Wolf C, Kretschmer S, Roers A. Type I interferonopathies—an expanding disease spectrum of immunodysregulation. Semin Immunopathol. 2015;37(4):349–57.CrossRef Lee-Kirsch MA, Wolf C, Kretschmer S, Roers A. Type I interferonopathies—an expanding disease spectrum of immunodysregulation. Semin Immunopathol. 2015;37(4):349–57.CrossRef
35.
go back to reference Genovese MC, Kremer JM, Kartman CE, Schlichting DE, Xie L, Carmack T, et al. Response to baricitinib based on prior biologic use in patients with refractory rheumatoid arthritis. Rheumatology. 2018;374:1243–9. Genovese MC, Kremer JM, Kartman CE, Schlichting DE, Xie L, Carmack T, et al. Response to baricitinib based on prior biologic use in patients with refractory rheumatoid arthritis. Rheumatology. 2018;374:1243–9.
36.
go back to reference Genovese MC, Greenwald M, Codding C, Zubrzycka-Sienkiewicz A, Kivitz AJ, Wang A, et al. Peficitinib, a JAK inhibitor, in combination with limited conventional synthetic disease-modifying antirheumatic drugs in the treatment of moderate-to-severe rheumatoid arthritis. Arthritis Rheumatol. 2017;69(5):932–42.CrossRef Genovese MC, Greenwald M, Codding C, Zubrzycka-Sienkiewicz A, Kivitz AJ, Wang A, et al. Peficitinib, a JAK inhibitor, in combination with limited conventional synthetic disease-modifying antirheumatic drugs in the treatment of moderate-to-severe rheumatoid arthritis. Arthritis Rheumatol. 2017;69(5):932–42.CrossRef
37.
go back to reference Dhillon S. Tofacitinib: a review in rheumatoid arthritis. Drugs. Springer International Publishing; 2017;:1–15. Dhillon S. Tofacitinib: a review in rheumatoid arthritis. Drugs. Springer International Publishing; 2017;:1–15.
38.
go back to reference Lee EB, Fleischmann R, Hall S, Wilkinson B, Bradley JD, Gruben D, et al. Tofacitinib versus methotrexate in rheumatoid arthritis. N Engl J Med. 2014;370(25):2377–86.CrossRef Lee EB, Fleischmann R, Hall S, Wilkinson B, Bradley JD, Gruben D, et al. Tofacitinib versus methotrexate in rheumatoid arthritis. N Engl J Med. 2014;370(25):2377–86.CrossRef
39.
go back to reference van Vollenhoven RF, Fleischmann R, Cohen S, Lee EB, García Meijide JA, Wagner S, et al. Tofacitinib or adalimumab versus placebo in rheumatoid arthritis. N Engl J Med. 2012;367(6):508–19.CrossRef van Vollenhoven RF, Fleischmann R, Cohen S, Lee EB, García Meijide JA, Wagner S, et al. Tofacitinib or adalimumab versus placebo in rheumatoid arthritis. N Engl J Med. 2012;367(6):508–19.CrossRef
40.
go back to reference Mease P, Hall S, FitzGerald O, van der Heijde D, Merola JF, Avila-Zapata F, et al. Tofacitinib or adalimumab versus placebo for psoriatic arthritis. N Engl J Med. 2017;377(16):1537–50.CrossRef Mease P, Hall S, FitzGerald O, van der Heijde D, Merola JF, Avila-Zapata F, et al. Tofacitinib or adalimumab versus placebo for psoriatic arthritis. N Engl J Med. 2017;377(16):1537–50.CrossRef
41.
go back to reference Gladman D, Rigby W, Azevedo VF, Behrens F, Blanco R, Kaszuba A, et al. Tofacitinib for psoriatic arthritis in patients with an inadequate response to TNF inhibitors. N Engl J Med. 2017;377(16):1525–36.CrossRef Gladman D, Rigby W, Azevedo VF, Behrens F, Blanco R, Kaszuba A, et al. Tofacitinib for psoriatic arthritis in patients with an inadequate response to TNF inhibitors. N Engl J Med. 2017;377(16):1525–36.CrossRef
42.
go back to reference Ruperto N, Brunner HI, Zuber Z, Tzaribachev N, Kingsbury DJ, 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;:1–10. Ruperto N, Brunner HI, Zuber Z, Tzaribachev N, Kingsbury DJ, 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;:1–10.
43.
go back to reference 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.CrossRef 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.CrossRef
44.
go back to reference Rodero MP, Frémond M-L, Rice GI, Neven B, Crow YJ. JAK inhibition in STING-associated interferonopathy. Ann Rheum Dis 2016;75(12):e75–5. Rodero MP, Frémond M-L, Rice GI, Neven B, Crow YJ. JAK inhibition in STING-associated interferonopathy. Ann Rheum Dis 2016;75(12):e75–5.
45.
go back to reference Seo J, Kang J-A, MD DIS, BS E-BP, MS C-RL, MD SAC, 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. Elsevier Ltd; 2017;139(4):1396–1399.e12. Seo J, Kang J-A, MD DIS, BS E-BP, MS C-RL, MD SAC, 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. Elsevier Ltd; 2017;139(4):1396–1399.e12.
46.
go back to reference Kuriya B, Cohen MD, Keystone E. Baricitinib in rheumatoid arthritis: evidence-to-date and clinical potential. Therapeutic Advances in Musculoskeletal. SAGE PublicationsSage UK: London, England; 2017;9(2):37–44. Kuriya B, Cohen MD, Keystone E. Baricitinib in rheumatoid arthritis: evidence-to-date and clinical potential. Therapeutic Advances in Musculoskeletal. SAGE PublicationsSage UK: London, England; 2017;9(2):37–44.
47.
go back to reference Taylor PC, Azeez MA, Kiriakidis S. Filgotinib for the treatment of rheumatoid arthritis. Expert Opin Investig Drugs. Taylor & Francis; 2017;00(00):1–7. Taylor PC, Azeez MA, Kiriakidis S. Filgotinib for the treatment of rheumatoid arthritis. Expert Opin Investig Drugs. Taylor & Francis; 2017;00(00):1–7.
48.
go back to reference Genovese MC, Kremer J, Zamani O, Ludivico C, Krogulec M, Xie L, et al. Baricitinib in Patients with Refractory Rheumatoid Arthritis. N Engl J Med. 2016;374(13):1243–52.CrossRef Genovese MC, Kremer J, Zamani O, Ludivico C, Krogulec M, Xie L, et al. Baricitinib in Patients with Refractory Rheumatoid Arthritis. N Engl J Med. 2016;374(13):1243–52.CrossRef
49.
go back to reference Sanchez GAM, Reinhardt A, Ramsey S, Wittkowski H, Hashkes PJ, Berkun Y, et al. JAK1/2 inhibition with baricitinib in the treatment of autoinflammatory interferonopathies. J Clin Investig. 2018;:1–30. Sanchez GAM, Reinhardt A, Ramsey S, Wittkowski H, Hashkes PJ, Berkun Y, et al. JAK1/2 inhibition with baricitinib in the treatment of autoinflammatory interferonopathies. J Clin Investig. 2018;:1–30.
50.
go back to reference Vincenti F, Tedesco Silva H, Busque S, O’Connell P, Friedewald J, Cibrik D, et al. Randomized phase 2b trial of tofacitinib (CP-690,550) in de novo kidney transplant patients: efficacy, renal function and safety at 1 year. Am J Transplant. Wiley/Blackwell (10.1111); 2012;12(9):2446–56. Vincenti F, Tedesco Silva H, Busque S, O’Connell P, Friedewald J, Cibrik D, et al. Randomized phase 2b trial of tofacitinib (CP-690,550) in de novo kidney transplant patients: efficacy, renal function and safety at 1 year. Am J Transplant. Wiley/Blackwell (10.1111); 2012;12(9):2446–56.
51.
go back to reference Frémond M-L, Uggenti C, Van Eyck L, Melki I, Bondet V, Kitabayashi N, et al. Brief report: blockade of TANK-binding kinase 1/IKKɛ inhibits mutant stimulator of interferon genes (STING)-mediated inflammatory responses in human peripheral blood mononuclear cells. Arthritis & Rheumatology. 2017;69(7):1495–501.CrossRef Frémond M-L, Uggenti C, Van Eyck L, Melki I, Bondet V, Kitabayashi N, et al. Brief report: blockade of TANK-binding kinase 1/IKKɛ inhibits mutant stimulator of interferon genes (STING)-mediated inflammatory responses in human peripheral blood mononuclear cells. Arthritis & Rheumatology. 2017;69(7):1495–501.CrossRef
52.
go back to reference Cohen SB, Tanaka Y, Mariette X, Curtis JR, Lee EB, Nash P, et al. Long-term safety of tofacitinib for the treatment of rheumatoid arthritis up to 8.5 years: integrated analysis of data from the global clinical trials. Ann Rheum Dis. 2017;76(7):1253–62.CrossRef Cohen SB, Tanaka Y, Mariette X, Curtis JR, Lee EB, Nash P, et al. Long-term safety of tofacitinib for the treatment of rheumatoid arthritis up to 8.5 years: integrated analysis of data from the global clinical trials. Ann Rheum Dis. 2017;76(7):1253–62.CrossRef
53.
go back to reference Winthrop KL. The emerging safety profile of JAK inhibitors in rheumatic disease. Nat Rev Rheumatol. 2017;13(4):234–43.CrossRef Winthrop KL. The emerging safety profile of JAK inhibitors in rheumatic disease. Nat Rev Rheumatol. 2017;13(4):234–43.CrossRef
54.
go back to reference Wollenhaupt J, Silverfield J, Lee EB, Curtis JR, Wood SP, Soma K, et al. Safety and efficacy of tofacitinib, an oral janus kinase inhibitor, for the treatment of rheumatoid arthritis in open-label, longterm extension studies. J Rheumatol. 2014;41(5):837–52.CrossRef Wollenhaupt J, Silverfield J, Lee EB, Curtis JR, Wood SP, Soma K, et al. Safety and efficacy of tofacitinib, an oral janus kinase inhibitor, for the treatment of rheumatoid arthritis in open-label, longterm extension studies. J Rheumatol. 2014;41(5):837–52.CrossRef
55.
go back to reference Avina-Zubieta JA, Thomas J, Sadatsafavi M, Lehman AJ, Lacaille D. Risk of incident cardiovascular events in patients with rheumatoid arthritis: a meta-analysis of observational studies. Ann Rheum Dis. 2012;71(9):1524–9.CrossRef Avina-Zubieta JA, Thomas J, Sadatsafavi M, Lehman AJ, Lacaille D. Risk of incident cardiovascular events in patients with rheumatoid arthritis: a meta-analysis of observational studies. Ann Rheum Dis. 2012;71(9):1524–9.CrossRef
56.
go back to reference Charles-Schoeman C, Wicker P, Gonzalez-Gay MA, Boy M, Zuckerman A, Soma K, et al. Cardiovascular safety findings in patients with rheumatoid arthritis treated with tofacitinib, an oral Janus kinase inhibitor. Semin Arthritis Rheum. Elsevier; 2016;46(3):261–71. Charles-Schoeman C, Wicker P, Gonzalez-Gay MA, Boy M, Zuckerman A, Soma K, et al. Cardiovascular safety findings in patients with rheumatoid arthritis treated with tofacitinib, an oral Janus kinase inhibitor. Semin Arthritis Rheum. Elsevier; 2016;46(3):261–71.
57.
go back to reference He Y, Wong AY, Chan EW, Lau WC, Man KK, Chui CS, et al. Efficacy and safety of tofacitinib in the treatment of rheumatoid arthritis: a systematic review and meta-analysis. BMC Musculoskelet Disord. BioMed Central; 2013;14(1):936. He Y, Wong AY, Chan EW, Lau WC, Man KK, Chui CS, et al. Efficacy and safety of tofacitinib in the treatment of rheumatoid arthritis: a systematic review and meta-analysis. BMC Musculoskelet Disord. BioMed Central; 2013;14(1):936.
58.
go back to reference Petty RE, Southwood TR, Manners P, Baum J, Glass DN, Goldenberg J, et al. International League of Associations for Rheumatology classification of juvenile idiopathic arthritis: second revision, Edmonton, 2001. J Rheumatol. 2004;31:390–2. Petty RE, Southwood TR, Manners P, Baum J, Glass DN, Goldenberg J, et al. International League of Associations for Rheumatology classification of juvenile idiopathic arthritis: second revision, Edmonton, 2001. J Rheumatol. 2004;31:390–2.
Metadata
Title
JAK Inhibitors in Rheumatology: Implications for Paediatric Syndromes?
Authors
S. A. Kerrigan
I. B. McInnes
Publication date
01-12-2018
Publisher
Springer US
Published in
Current Rheumatology Reports / Issue 12/2018
Print ISSN: 1523-3774
Electronic ISSN: 1534-6307
DOI
https://doi.org/10.1007/s11926-018-0792-7

Other articles of this Issue 12/2018

Current Rheumatology Reports 12/2018 Go to the issue

Orphan Diseases (B Manger, Section Editor)

Chronic Periaortitis: an Update

Complementary and Alternative Medicine (S Kolasinski, Section Editor)

Systematic Review and Meta-analysis: Mindfulness-Based Interventions for Rheumatoid Arthritis

Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.