Skip to main content
Erschienen in: Clinical Reviews in Allergy & Immunology 2/2020

17.06.2020

Type I Interferons in the Pathogenesis and Treatment of Autoimmune Diseases

verfasst von: Jiao Jiang, Ming Zhao, Christopher Chang, Haijing Wu, Qianjin Lu

Erschienen in: Clinical Reviews in Allergy & Immunology | Ausgabe 2/2020

Einloggen, um Zugang zu erhalten

Abstract

Type I interferons (IFN-Is) are a very important group of cytokines that are produced by innate immune cells but also act on adaptive immune cells. IFN-Is possess antiviral, antitumor, and anti-proliferative effects, as well are associated with the initiation and maintenance of autoimmune disorders. Studies have shown that aberrantly expressed IFN-Is and/or type I IFN-inducible gene signatures in the serum or tissues of patients with autoimmune disorders are linked to their pathogenesis, clinical manifestations, and disease activity. Type I interferonopathies with mutations in genes impacting the type I IFN signaling pathway have shown symptoms and characteristics similar to those of systemic lupus erythematosus (SLE). Furthermore, both interventions in animal models and clinical trials of therapies targeting the type I IFN signaling pathway have shown efficacy in the treatment of autoimmune diseases. Our review aims to summarize the functions and targeted therapies (as well as clinical trials) of IFN-Is in both adult and pediatric autoimmune diseases, such as SLE, pediatric SLE (pSLE), rheumatoid arthritis (RA), juvenile idiopathic arthritis (JIA), juvenile dermatomyositis (JDM), Sjögren syndrome (SjS), and systemic sclerosis (SSc), discussing the potential abnormal regulation of transcription factors and epigenetic modifications and providing a potential mechanism for pathogenesis and therapeutic strategies for future clinical use.
Literatur
13.
Zurück zum Zitat Higgs BW, Liu Z, White B, Zhu W, White WI, Morehouse C, Brohawn P, Kiener PA, Richman L, Fiorentino D, Greenberg SA, Jallal B, Yao Y (2011) Patients with systemic lupus erythematosus, myositis, rheumatoid arthritis and scleroderma share activation of a common type I interferon pathway. Ann Rheum Dis 70(11):2029–2036. https://doi.org/10.1136/ard.2011.150326CrossRefPubMed Higgs BW, Liu Z, White B, Zhu W, White WI, Morehouse C, Brohawn P, Kiener PA, Richman L, Fiorentino D, Greenberg SA, Jallal B, Yao Y (2011) Patients with systemic lupus erythematosus, myositis, rheumatoid arthritis and scleroderma share activation of a common type I interferon pathway. Ann Rheum Dis 70(11):2029–2036. https://​doi.​org/​10.​1136/​ard.​2011.​150326CrossRefPubMed
23.
Zurück zum Zitat Kato Y, Park J, Takamatsu H, Konaka H, Aoki W, Aburaya S, Ueda M, Nishide M, Koyama S, Hayama Y, Kinehara Y, Hirano T, Shima Y, Narazaki M, Kumanogoh A (2018) Apoptosis-derived membrane vesicles drive the cGAS-STING pathway and enhance type I IFN production in systemic lupus erythematosus. Ann Rheum Dis 77(10):1507–1515. https://doi.org/10.1136/annrheumdis-2018-212988CrossRefPubMed Kato Y, Park J, Takamatsu H, Konaka H, Aoki W, Aburaya S, Ueda M, Nishide M, Koyama S, Hayama Y, Kinehara Y, Hirano T, Shima Y, Narazaki M, Kumanogoh A (2018) Apoptosis-derived membrane vesicles drive the cGAS-STING pathway and enhance type I IFN production in systemic lupus erythematosus. Ann Rheum Dis 77(10):1507–1515. https://​doi.​org/​10.​1136/​annrheumdis-2018-212988CrossRefPubMed
31.
Zurück zum Zitat Jonsson KL, Laustsen A, Krapp C, Skipper KA, Thavachelvam K, Hotter D, Egedal JH, Kjolby M, Mohammadi P, Prabakaran T, Sorensen LK, Sun C, Jensen SB, Holm CK, Lebbink RJ, Johannsen M, Nyegaard M, Mikkelsen JG, Kirchhoff F, Paludan SR, Jakobsen MR (2017) IFI16 is required for DNA sensing in human macrophages by promoting production and function of cGAMP. Nat Commun 8:14391. https://doi.org/10.1038/ncomms14391CrossRefPubMedPubMedCentral Jonsson KL, Laustsen A, Krapp C, Skipper KA, Thavachelvam K, Hotter D, Egedal JH, Kjolby M, Mohammadi P, Prabakaran T, Sorensen LK, Sun C, Jensen SB, Holm CK, Lebbink RJ, Johannsen M, Nyegaard M, Mikkelsen JG, Kirchhoff F, Paludan SR, Jakobsen MR (2017) IFI16 is required for DNA sensing in human macrophages by promoting production and function of cGAMP. Nat Commun 8:14391. https://​doi.​org/​10.​1038/​ncomms14391CrossRefPubMedPubMedCentral
47.
62.
73.
Zurück zum Zitat Imgenberg-Kreuz J, Carlsson Almlof J, Leonard D, Alexsson A, Nordmark G, Eloranta ML, Rantapaa-Dahlqvist S, Bengtsson AA, Jonsen A, Padyukov L, Gunnarsson I, Svenungsson E, Sjowall C, Ronnblom L, Syvanen AC, Sandling JK (2018) DNA methylation mapping identifies gene regulatory effects in patients with systemic lupus erythematosus. Ann Rheum Dis 77(5):736–743. https://doi.org/10.1136/annrheumdis-2017-212379CrossRefPubMed Imgenberg-Kreuz J, Carlsson Almlof J, Leonard D, Alexsson A, Nordmark G, Eloranta ML, Rantapaa-Dahlqvist S, Bengtsson AA, Jonsen A, Padyukov L, Gunnarsson I, Svenungsson E, Sjowall C, Ronnblom L, Syvanen AC, Sandling JK (2018) DNA methylation mapping identifies gene regulatory effects in patients with systemic lupus erythematosus. Ann Rheum Dis 77(5):736–743. https://​doi.​org/​10.​1136/​annrheumdis-2017-212379CrossRefPubMed
75.
Zurück zum Zitat Imgenberg-Kreuz J, Sandling JK, Almlof JC, Nordlund J, Signer L, Norheim KB, Omdal R, Ronnblom L, Eloranta ML, Syvanen AC, Nordmark G (2016) Genome-wide DNA methylation analysis in multiple tissues in primary Sjogren’s syndrome reveals regulatory effects at interferon-induced genes. Ann Rheum Dis 75(11):2029–2036. https://doi.org/10.1136/annrheumdis-2015-208659CrossRefPubMed Imgenberg-Kreuz J, Sandling JK, Almlof JC, Nordlund J, Signer L, Norheim KB, Omdal R, Ronnblom L, Eloranta ML, Syvanen AC, Nordmark G (2016) Genome-wide DNA methylation analysis in multiple tissues in primary Sjogren’s syndrome reveals regulatory effects at interferon-induced genes. Ann Rheum Dis 75(11):2029–2036. https://​doi.​org/​10.​1136/​annrheumdis-2015-208659CrossRefPubMed
76.
Zurück zum Zitat Ding W, Pu W, Wang L, Jiang S, Zhou X, Tu W, Yu L, Zhang J, Guo S, Liu Q, Ma Y, Chen S, Wu W, Reveille J, Zou H, Jin L, Wang J (2018) Genome-wide DNA methylation analysis in systemic sclerosis reveals hypomethylation of IFN-associated genes in CD4(+) and CD8(+) T cells. J Investig Dermatol 138(5):1069–1077. https://doi.org/10.1016/j.jid.2017.12.003CrossRefPubMed Ding W, Pu W, Wang L, Jiang S, Zhou X, Tu W, Yu L, Zhang J, Guo S, Liu Q, Ma Y, Chen S, Wu W, Reveille J, Zou H, Jin L, Wang J (2018) Genome-wide DNA methylation analysis in systemic sclerosis reveals hypomethylation of IFN-associated genes in CD4(+) and CD8(+) T cells. J Investig Dermatol 138(5):1069–1077. https://​doi.​org/​10.​1016/​j.​jid.​2017.​12.​003CrossRefPubMed
77.
86.
Zurück zum Zitat Decque A, Joffre O, Magalhaes JG, Cossec JC, Blecher-Gonen R, Lapaquette P, Silvin A, Manel N, Joubert PE, Seeler JS, Albert ML, Amit I, Amigorena S, Dejean A (2016) Sumoylation coordinates the repression of inflammatory and anti-viral gene-expression programs during innate sensing. Nat Immunol 17(2):140–149. https://doi.org/10.1038/ni.3342CrossRefPubMed Decque A, Joffre O, Magalhaes JG, Cossec JC, Blecher-Gonen R, Lapaquette P, Silvin A, Manel N, Joubert PE, Seeler JS, Albert ML, Amit I, Amigorena S, Dejean A (2016) Sumoylation coordinates the repression of inflammatory and anti-viral gene-expression programs during innate sensing. Nat Immunol 17(2):140–149. https://​doi.​org/​10.​1038/​ni.​3342CrossRefPubMed
87.
Zurück zum Zitat Zhang Y, Mao D, Roswit WT, Jin X, Patel AC, Patel DA, Agapov E, Wang Z, Tidwell RM, Atkinson JJ, Huang G, McCarthy R, Yu J, Yun NE, Paessler S, Lawson TG, Omattage NS, Brett TJ, Holtzman MJ (2015) PARP9-DTX3L ubiquitin ligase targets host histone H2BJ and viral 3C protease to enhance interferon signaling and control viral infection. Nat Immunol 16(12):1215–1227. https://doi.org/10.1038/ni.3279CrossRefPubMedPubMedCentral Zhang Y, Mao D, Roswit WT, Jin X, Patel AC, Patel DA, Agapov E, Wang Z, Tidwell RM, Atkinson JJ, Huang G, McCarthy R, Yu J, Yun NE, Paessler S, Lawson TG, Omattage NS, Brett TJ, Holtzman MJ (2015) PARP9-DTX3L ubiquitin ligase targets host histone H2BJ and viral 3C protease to enhance interferon signaling and control viral infection. Nat Immunol 16(12):1215–1227. https://​doi.​org/​10.​1038/​ni.​3279CrossRefPubMedPubMedCentral
90.
Zurück zum Zitat Han X, Wang Y, Zhang X, Qin Y, Qu B, Wu L, Ma J, Zhou Z, Qian J, Dai M, Tang Y, Chan EK, Harley JB, Zhou S, Shen N (2016) MicroRNA-130b ameliorates murine lupus nephritis through targeting the type I interferon pathway on renal mesangial cells. Arthritis Rheumatol (Hoboken, NJ) 68(9):2232–2243. https://doi.org/10.1002/art.39725CrossRef Han X, Wang Y, Zhang X, Qin Y, Qu B, Wu L, Ma J, Zhou Z, Qian J, Dai M, Tang Y, Chan EK, Harley JB, Zhou S, Shen N (2016) MicroRNA-130b ameliorates murine lupus nephritis through targeting the type I interferon pathway on renal mesangial cells. Arthritis Rheumatol (Hoboken, NJ) 68(9):2232–2243. https://​doi.​org/​10.​1002/​art.​39725CrossRef
91.
Zurück zum Zitat Rossato M, Affandi AJ, Thordardottir S, Wichers CGK, Cossu M, Broen JCA, Moret FM, Bossini-Castillo L, Chouri E, van Bon L, Wolters F, Marut W, van der Kroef M, Silva-Cardoso S, Bekker CPJ, Dolstra H, van Laar JM, Martin J, van Roon JAG, Reedquist KA, Beretta L, Radstake T (2017) Association of MicroRNA-618 expression with altered frequency and activation of plasmacytoid dendritic cells in patients with systemic sclerosis. Arthritis Rheumatol (Hoboken, NJ) 69(9):1891–1902. https://doi.org/10.1002/art.40163CrossRef Rossato M, Affandi AJ, Thordardottir S, Wichers CGK, Cossu M, Broen JCA, Moret FM, Bossini-Castillo L, Chouri E, van Bon L, Wolters F, Marut W, van der Kroef M, Silva-Cardoso S, Bekker CPJ, Dolstra H, van Laar JM, Martin J, van Roon JAG, Reedquist KA, Beretta L, Radstake T (2017) Association of MicroRNA-618 expression with altered frequency and activation of plasmacytoid dendritic cells in patients with systemic sclerosis. Arthritis Rheumatol (Hoboken, NJ) 69(9):1891–1902. https://​doi.​org/​10.​1002/​art.​40163CrossRef
92.
93.
Zurück zum Zitat Smith S, Fernando T, Wu PW, Seo J, Ni Gabhann J, Piskareva O, McCarthy E, Howard D, O'Connell P, Conway R, Gallagher P, Molloy E, Stallings RL, Kearns G, Forbess L, Ishimori M, Venuturupalli S, Wallace D, Weisman M, Jefferies CA (2017) MicroRNA-302d targets IRF9 to regulate the IFN-induced gene expression in SLE. J Autoimmun 79:105–111. https://doi.org/10.1016/j.jaut.2017.03.003CrossRefPubMed Smith S, Fernando T, Wu PW, Seo J, Ni Gabhann J, Piskareva O, McCarthy E, Howard D, O'Connell P, Conway R, Gallagher P, Molloy E, Stallings RL, Kearns G, Forbess L, Ishimori M, Venuturupalli S, Wallace D, Weisman M, Jefferies CA (2017) MicroRNA-302d targets IRF9 to regulate the IFN-induced gene expression in SLE. J Autoimmun 79:105–111. https://​doi.​org/​10.​1016/​j.​jaut.​2017.​03.​003CrossRefPubMed
97.
106.
107.
Zurück zum Zitat Jacob CO, Zhu J, Armstrong DL, Yan M, Han J, Zhou XJ, Thomas JA, Reiff A, Myones BL, Ojwang JO, Kaufman KM, Klein-Gitelman M, McCurdy D, Wagner-Weiner L, Silverman E, Ziegler J, Kelly JA, Merrill JT, Harley JB, Ramsey-Goldman R, Vila LM, Bae SC, Vyse TJ, Gilkeson GS, Gaffney PM, Moser KL, Langefeld CD, Zidovetzki R, Mohan C (2009) Identification of IRAK1 as a risk gene with critical role in the pathogenesis of systemic lupus erythematosus. Proc Natl Acad Sci U S A 106(15):6256–6261. https://doi.org/10.1073/pnas.0901181106CrossRefPubMedPubMedCentral Jacob CO, Zhu J, Armstrong DL, Yan M, Han J, Zhou XJ, Thomas JA, Reiff A, Myones BL, Ojwang JO, Kaufman KM, Klein-Gitelman M, McCurdy D, Wagner-Weiner L, Silverman E, Ziegler J, Kelly JA, Merrill JT, Harley JB, Ramsey-Goldman R, Vila LM, Bae SC, Vyse TJ, Gilkeson GS, Gaffney PM, Moser KL, Langefeld CD, Zidovetzki R, Mohan C (2009) Identification of IRAK1 as a risk gene with critical role in the pathogenesis of systemic lupus erythematosus. Proc Natl Acad Sci U S A 106(15):6256–6261. https://​doi.​org/​10.​1073/​pnas.​0901181106CrossRefPubMedPubMedCentral
111.
Zurück zum Zitat Wang Y, Shaked I, Stanford SM, Zhou W, Curtsinger JM, Mikulski Z, Shaheen ZR, Cheng G, Sawatzke K, Campbell AM, Auger JL, Bilgic H, Shoyama FM, Schmeling DO, Balfour HH Jr, Hasegawa K, Chan AC, Corbett JA, Binstadt BA, Mescher MF, Ley K, Bottini N, Peterson EJ (2013) The autoimmunity-associated gene PTPN22 potentiates toll-like receptor-driven, type 1 interferon-dependent immunity. Immunity 39(1):111–122. https://doi.org/10.1016/j.immuni.2013.06.013CrossRefPubMed Wang Y, Shaked I, Stanford SM, Zhou W, Curtsinger JM, Mikulski Z, Shaheen ZR, Cheng G, Sawatzke K, Campbell AM, Auger JL, Bilgic H, Shoyama FM, Schmeling DO, Balfour HH Jr, Hasegawa K, Chan AC, Corbett JA, Binstadt BA, Mescher MF, Ley K, Bottini N, Peterson EJ (2013) The autoimmunity-associated gene PTPN22 potentiates toll-like receptor-driven, type 1 interferon-dependent immunity. Immunity 39(1):111–122. https://​doi.​org/​10.​1016/​j.​immuni.​2013.​06.​013CrossRefPubMed
114.
Zurück zum Zitat Hinks A, Cobb J, Marion MC, Prahalad S, Sudman M, Bowes J, Martin P, Comeau ME, Sajuthi S, Andrews R, Brown M, Chen WM, Concannon P, Deloukas P, Edkins S, Eyre S, Gaffney PM, Guthery SL, Guthridge JM, Hunt SE, James JA, Keddache M, Moser KL, Nigrovic PA, Onengut-Gumuscu S, Onslow ML, Rose CD, Rich SS, Steel KJ, Wakeland EK, Wallace CA, Wedderburn LR, Woo P, Boston Children's JIAR, British Society of P, Adolescent Rheumatology Study G, Childhood Arthritis Prospective S, Childhood Arthritis Response to Medication S, German Society for Pediatric R, Study JIAGE, Registry NJG, Study T, United Kingdom Juvenile Idiopathic Arthritis Genetics C, Bohnsack JF, Haas JP, Glass DN, Langefeld CD, Thomson W, Thompson SD (2013) Dense genotyping of immune-related disease regions identifies 14 new susceptibility loci for juvenile idiopathic arthritis. Nat Genet 45(6):664–669. https://doi.org/10.1038/ng.2614CrossRefPubMedPubMedCentral Hinks A, Cobb J, Marion MC, Prahalad S, Sudman M, Bowes J, Martin P, Comeau ME, Sajuthi S, Andrews R, Brown M, Chen WM, Concannon P, Deloukas P, Edkins S, Eyre S, Gaffney PM, Guthery SL, Guthridge JM, Hunt SE, James JA, Keddache M, Moser KL, Nigrovic PA, Onengut-Gumuscu S, Onslow ML, Rose CD, Rich SS, Steel KJ, Wakeland EK, Wallace CA, Wedderburn LR, Woo P, Boston Children's JIAR, British Society of P, Adolescent Rheumatology Study G, Childhood Arthritis Prospective S, Childhood Arthritis Response to Medication S, German Society for Pediatric R, Study JIAGE, Registry NJG, Study T, United Kingdom Juvenile Idiopathic Arthritis Genetics C, Bohnsack JF, Haas JP, Glass DN, Langefeld CD, Thomson W, Thompson SD (2013) Dense genotyping of immune-related disease regions identifies 14 new susceptibility loci for juvenile idiopathic arthritis. Nat Genet 45(6):664–669. https://​doi.​org/​10.​1038/​ng.​2614CrossRefPubMedPubMedCentral
115.
Zurück zum Zitat Nordang GB, Viken MK, Amundsen SS, Sanchez ES, Flato B, Forre OT, Martin J, Kvien TK, Lie BA (2012) Interferon regulatory factor 5 gene polymorphism confers risk to several rheumatic diseases and correlates with expression of alternative thymic transcripts. Rheumatology (Oxford, England) 51(4):619–626. https://doi.org/10.1093/rheumatology/ker364CrossRef Nordang GB, Viken MK, Amundsen SS, Sanchez ES, Flato B, Forre OT, Martin J, Kvien TK, Lie BA (2012) Interferon regulatory factor 5 gene polymorphism confers risk to several rheumatic diseases and correlates with expression of alternative thymic transcripts. Rheumatology (Oxford, England) 51(4):619–626. https://​doi.​org/​10.​1093/​rheumatology/​ker364CrossRef
116.
Zurück zum Zitat Harris VM, Scofield RH, Sivils KL (2019) Genetics in Sjogren’s syndrome: where we are and where we go. Clin Exp Rheumatol 37 Suppl 118(3):234–239PubMed Harris VM, Scofield RH, Sivils KL (2019) Genetics in Sjogren’s syndrome: where we are and where we go. Clin Exp Rheumatol 37 Suppl 118(3):234–239PubMed
120.
Zurück zum Zitat Crow YJ, Hayward BE, Parmar R, Robins P, Leitch A, Ali M, Black DN, van Bokhoven H, Brunner HG, Hamel BC, Corry PC, Cowan FM, Frints SG, Klepper J, Livingston JH, Lynch SA, Massey RF, Meritet JF, Michaud JL, Ponsot G, Voit T, Lebon P, Bonthron DT, Jackson AP, Barnes DE, Lindahl T (2006) Mutations in the gene encoding the 3′-5' DNA exonuclease TREX1 cause Aicardi-Goutieres syndrome at the AGS1 locus. Nat Genet 38(8):917–920. https://doi.org/10.1038/ng1845CrossRefPubMed Crow YJ, Hayward BE, Parmar R, Robins P, Leitch A, Ali M, Black DN, van Bokhoven H, Brunner HG, Hamel BC, Corry PC, Cowan FM, Frints SG, Klepper J, Livingston JH, Lynch SA, Massey RF, Meritet JF, Michaud JL, Ponsot G, Voit T, Lebon P, Bonthron DT, Jackson AP, Barnes DE, Lindahl T (2006) Mutations in the gene encoding the 3′-5' DNA exonuclease TREX1 cause Aicardi-Goutieres syndrome at the AGS1 locus. Nat Genet 38(8):917–920. https://​doi.​org/​10.​1038/​ng1845CrossRefPubMed
121.
Zurück zum Zitat Crow YJ, Leitch A, Hayward BE, Garner A, Parmar R, Griffith E, Ali M, Semple C, Aicardi J, Babul-Hirji R, Baumann C, Baxter P, Bertini E, Chandler KE, Chitayat D, Cau D, Dery C, Fazzi E, Goizet C, King MD, Klepper J, Lacombe D, Lanzi G, Lyall H, Martinez-Frias ML, Mathieu M, McKeown C, Monier A, Oade Y, Quarrell OW, Rittey CD, Rogers RC, Sanchis A, Stephenson JB, Tacke U, Till M, Tolmie JL, Tomlin P, Voit T, Weschke B, Woods CG, Lebon P, Bonthron DT, Ponting CP, Jackson AP (2006) Mutations in genes encoding ribonuclease H2 subunits cause Aicardi-Goutieres syndrome and mimic congenital viral brain infection. Nat Genet 38(8):910–916. https://doi.org/10.1038/ng1842CrossRefPubMed Crow YJ, Leitch A, Hayward BE, Garner A, Parmar R, Griffith E, Ali M, Semple C, Aicardi J, Babul-Hirji R, Baumann C, Baxter P, Bertini E, Chandler KE, Chitayat D, Cau D, Dery C, Fazzi E, Goizet C, King MD, Klepper J, Lacombe D, Lanzi G, Lyall H, Martinez-Frias ML, Mathieu M, McKeown C, Monier A, Oade Y, Quarrell OW, Rittey CD, Rogers RC, Sanchis A, Stephenson JB, Tacke U, Till M, Tolmie JL, Tomlin P, Voit T, Weschke B, Woods CG, Lebon P, Bonthron DT, Ponting CP, Jackson AP (2006) Mutations in genes encoding ribonuclease H2 subunits cause Aicardi-Goutieres syndrome and mimic congenital viral brain infection. Nat Genet 38(8):910–916. https://​doi.​org/​10.​1038/​ng1842CrossRefPubMed
122.
Zurück zum Zitat Rice GI, Bond J, Asipu A, Brunette RL, Manfield IW, Carr IM, Fuller JC, Jackson RM, Lamb T, Briggs TA, Ali M, Gornall H, Couthard LR, Aeby A, Attard-Montalto SP, Bertini E, Bodemer C, Brockmann K, Brueton LA, Corry PC, Desguerre I, Fazzi E, Cazorla AG, Gener B, Hamel BC, Heiberg A, Hunter M, van der Knaap MS, Kumar R, Lagae L, Landrieu PG, Lourenco CM, Marom D, McDermott MF, van der Merwe W, Orcesi S, Prendiville JS, Rasmussen M, Shalev SA, Soler DM, Shinawi M, Spiegel R, Tan TY, Vanderver A, Wakeling EL, Wassmer E, Whittaker E, Lebon P, Stetson DB, Bonthron DT, Crow YJ (2009) Mutations involved in Aicardi-Goutieres syndrome implicate SAMHD1 as regulator of the innate immune response. Nat Genet 41(7):829–832. https://doi.org/10.1038/ng.373CrossRefPubMedPubMedCentral Rice GI, Bond J, Asipu A, Brunette RL, Manfield IW, Carr IM, Fuller JC, Jackson RM, Lamb T, Briggs TA, Ali M, Gornall H, Couthard LR, Aeby A, Attard-Montalto SP, Bertini E, Bodemer C, Brockmann K, Brueton LA, Corry PC, Desguerre I, Fazzi E, Cazorla AG, Gener B, Hamel BC, Heiberg A, Hunter M, van der Knaap MS, Kumar R, Lagae L, Landrieu PG, Lourenco CM, Marom D, McDermott MF, van der Merwe W, Orcesi S, Prendiville JS, Rasmussen M, Shalev SA, Soler DM, Shinawi M, Spiegel R, Tan TY, Vanderver A, Wakeling EL, Wassmer E, Whittaker E, Lebon P, Stetson DB, Bonthron DT, Crow YJ (2009) Mutations involved in Aicardi-Goutieres syndrome implicate SAMHD1 as regulator of the innate immune response. Nat Genet 41(7):829–832. https://​doi.​org/​10.​1038/​ng.​373CrossRefPubMedPubMedCentral
124.
Zurück zum Zitat Rice GI, Kasher PR, Forte GM, Mannion NM, Greenwood SM, Szynkiewicz M, Dickerson JE, Bhaskar SS, Zampini M, Briggs TA, Jenkinson EM, Bacino CA, Battini R, Bertini E, Brogan PA, Brueton LA, Carpanelli M, De Laet C, de Lonlay P, del Toro M, Desguerre I, Fazzi E, Garcia-Cazorla A, Heiberg A, Kawaguchi M, Kumar R, Lin JP, Lourenco CM, Male AM, Marques W Jr, Mignot C, Olivieri I, Orcesi S, Prabhakar P, Rasmussen M, Robinson RA, Rozenberg F, Schmidt JL, Steindl K, Tan TY, van der Merwe WG, Vanderver A, Vassallo G, Wakeling EL, Wassmer E, Whittaker E, Livingston JH, Lebon P, Suzuki T, McLaughlin PJ, Keegan LP, O'Connell MA, Lovell SC, Crow YJ (2012) Mutations in ADAR1 cause Aicardi-Goutieres syndrome associated with a type I interferon signature. Nat Genet 44(11):1243–1248. https://doi.org/10.1038/ng.2414CrossRefPubMedPubMedCentral Rice GI, Kasher PR, Forte GM, Mannion NM, Greenwood SM, Szynkiewicz M, Dickerson JE, Bhaskar SS, Zampini M, Briggs TA, Jenkinson EM, Bacino CA, Battini R, Bertini E, Brogan PA, Brueton LA, Carpanelli M, De Laet C, de Lonlay P, del Toro M, Desguerre I, Fazzi E, Garcia-Cazorla A, Heiberg A, Kawaguchi M, Kumar R, Lin JP, Lourenco CM, Male AM, Marques W Jr, Mignot C, Olivieri I, Orcesi S, Prabhakar P, Rasmussen M, Robinson RA, Rozenberg F, Schmidt JL, Steindl K, Tan TY, van der Merwe WG, Vanderver A, Vassallo G, Wakeling EL, Wassmer E, Whittaker E, Livingston JH, Lebon P, Suzuki T, McLaughlin PJ, Keegan LP, O'Connell MA, Lovell SC, Crow YJ (2012) Mutations in ADAR1 cause Aicardi-Goutieres syndrome associated with a type I interferon signature. Nat Genet 44(11):1243–1248. https://​doi.​org/​10.​1038/​ng.​2414CrossRefPubMedPubMedCentral
126.
Zurück zum Zitat Pelzer N, Hoogeveen ES, Haan J, Bunnik R, Poot CC, van Zwet EW, Inderson A, Fogteloo AJ, Reinders MEJ, Middelkoop HAM, Kruit MC, van den Maagdenberg A, Ferrari MD, Terwindt GM (2019) Systemic features of retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations: a monogenic small vessel disease. J Intern Med 285(3):317–332. https://doi.org/10.1111/joim.12848CrossRefPubMed Pelzer N, Hoogeveen ES, Haan J, Bunnik R, Poot CC, van Zwet EW, Inderson A, Fogteloo AJ, Reinders MEJ, Middelkoop HAM, Kruit MC, van den Maagdenberg A, Ferrari MD, Terwindt GM (2019) Systemic features of retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations: a monogenic small vessel disease. J Intern Med 285(3):317–332. https://​doi.​org/​10.​1111/​joim.​12848CrossRefPubMed
128.
Zurück zum Zitat Liu Y, Jesus AA, Marrero B, Yang D, Ramsey SE, Sanchez GAM, Tenbrock K, Wittkowski H, Jones OY, Kuehn HS, Lee CR, DiMattia MA, Cowen EW, Gonzalez B, Palmer I, DiGiovanna JJ, Biancotto A, Kim H, Tsai WL, Trier AM, Huang Y, Stone DL, Hill S, Kim HJ, St Hilaire C, Gurprasad S, Plass N, Chapelle D, Horkayne-Szakaly I, Foell D, Barysenka A, Candotti F, Holland SM, Hughes JD, Mehmet H, Issekutz AC, Raffeld M, McElwee J, Fontana JR, Minniti CP, Moir S, Kastner DL, Gadina M, Steven AC, Wingfield PT, Brooks SR, Rosenzweig SD, Fleisher TA, Deng Z, Boehm M, Paller AS, Goldbach-Mansky R (2014) Activated STING in a vascular and pulmonary syndrome. N Engl J Med 371(6):507–518. https://doi.org/10.1056/NEJMoa1312625CrossRefPubMedPubMedCentral Liu Y, Jesus AA, Marrero B, Yang D, Ramsey SE, Sanchez GAM, Tenbrock K, Wittkowski H, Jones OY, Kuehn HS, Lee CR, DiMattia MA, Cowen EW, Gonzalez B, Palmer I, DiGiovanna JJ, Biancotto A, Kim H, Tsai WL, Trier AM, Huang Y, Stone DL, Hill S, Kim HJ, St Hilaire C, Gurprasad S, Plass N, Chapelle D, Horkayne-Szakaly I, Foell D, Barysenka A, Candotti F, Holland SM, Hughes JD, Mehmet H, Issekutz AC, Raffeld M, McElwee J, Fontana JR, Minniti CP, Moir S, Kastner DL, Gadina M, Steven AC, Wingfield PT, Brooks SR, Rosenzweig SD, Fleisher TA, Deng Z, Boehm M, Paller AS, Goldbach-Mansky R (2014) Activated STING in a vascular and pulmonary syndrome. N Engl J Med 371(6):507–518. https://​doi.​org/​10.​1056/​NEJMoa1312625CrossRefPubMedPubMedCentral
135.
Zurück zum Zitat Brehm A, Liu Y, Sheikh A, Marrero B, Omoyinmi E, Zhou Q, Montealegre G, Biancotto A, Reinhardt A, Almeida de Jesus A, Pelletier M, Tsai WL, Remmers EF, Kardava L, Hill S, Kim H, Lachmann HJ, Megarbane A, Chae JJ, Brady J, Castillo RD, Brown D, Casano AV, Gao L, Chapelle D, Huang Y, Stone D, Chen Y, Sotzny F, Lee CC, Kastner DL, Torrelo A, Zlotogorski A, Moir S, Gadina M, McCoy P, Wesley R, Rother KI, Hildebrand PW, Brogan P, Kruger E, Aksentijevich I, Goldbach-Mansky R (2015) Additive loss-of-function proteasome subunit mutations in CANDLE/PRAAS patients promote type I IFN production. J Clin Invest 125(11):4196–4211. https://doi.org/10.1172/JCI81260CrossRefPubMedPubMedCentral Brehm A, Liu Y, Sheikh A, Marrero B, Omoyinmi E, Zhou Q, Montealegre G, Biancotto A, Reinhardt A, Almeida de Jesus A, Pelletier M, Tsai WL, Remmers EF, Kardava L, Hill S, Kim H, Lachmann HJ, Megarbane A, Chae JJ, Brady J, Castillo RD, Brown D, Casano AV, Gao L, Chapelle D, Huang Y, Stone D, Chen Y, Sotzny F, Lee CC, Kastner DL, Torrelo A, Zlotogorski A, Moir S, Gadina M, McCoy P, Wesley R, Rother KI, Hildebrand PW, Brogan P, Kruger E, Aksentijevich I, Goldbach-Mansky R (2015) Additive loss-of-function proteasome subunit mutations in CANDLE/PRAAS patients promote type I IFN production. J Clin Invest 125(11):4196–4211. https://​doi.​org/​10.​1172/​JCI81260CrossRefPubMedPubMedCentral
136.
Zurück zum Zitat Alsohime F, Martin-Fernandez M, Temsah MH, Alabdulhafid M, Le Voyer T, Alghamdi M, Qiu X, Alotaibi N, Alkahtani A, Buta S, Jouanguy E, Al-Eyadhy A, Gruber C, Hasan GM, Bashiri FA, Halwani R, Hassan HH, Al-Muhsen S, Alkhamis N, Alsum Z, Casanova JL, Bustamante J, Bogunovic D, Alangari AA (2020) JAK inhibitor therapy in a child with inherited USP18 deficiency. N Engl J Med 382(3):256–265. https://doi.org/10.1056/NEJMoa1905633CrossRefPubMedPubMedCentral Alsohime F, Martin-Fernandez M, Temsah MH, Alabdulhafid M, Le Voyer T, Alghamdi M, Qiu X, Alotaibi N, Alkahtani A, Buta S, Jouanguy E, Al-Eyadhy A, Gruber C, Hasan GM, Bashiri FA, Halwani R, Hassan HH, Al-Muhsen S, Alkhamis N, Alsum Z, Casanova JL, Bustamante J, Bogunovic D, Alangari AA (2020) JAK inhibitor therapy in a child with inherited USP18 deficiency. N Engl J Med 382(3):256–265. https://​doi.​org/​10.​1056/​NEJMoa1905633CrossRefPubMedPubMedCentral
137.
Zurück zum Zitat Sanchez GAM, Reinhardt A, Ramsey S, Wittkowski H, Hashkes PJ, Berkun Y, Schalm S, Murias S, Dare JA, Brown D, Stone DL, Gao L, Klausmeier T, Foell D, de Jesus AA, Chapelle DC, Kim H, Dill S, Colbert RA, Failla L, Kost B, O'Brien M, Reynolds JC, Folio LR, Calvo KR, Paul SM, Weir N, Brofferio A, Soldatos A, Biancotto A, Cowen EW, Digiovanna JJ, Gadina M, Lipton AJ, Hadigan C, Holland SM, Fontana J, Alawad AS, Brown RJ, Rother KI, Heller T, Brooks KM, Kumar P, Brooks SR, Waldman M, Singh HK, Nickeleit V, Silk M, Prakash A, Janes JM, Ozen S, Wakim PG, Brogan PA, Macias WL, Goldbach-Mansky R (2018) JAK1/2 inhibition with baricitinib in the treatment of autoinflammatory interferonopathies. J Clin Invest 128(7):3041–3052. https://doi.org/10.1172/JCI98814CrossRefPubMedPubMedCentral Sanchez GAM, Reinhardt A, Ramsey S, Wittkowski H, Hashkes PJ, Berkun Y, Schalm S, Murias S, Dare JA, Brown D, Stone DL, Gao L, Klausmeier T, Foell D, de Jesus AA, Chapelle DC, Kim H, Dill S, Colbert RA, Failla L, Kost B, O'Brien M, Reynolds JC, Folio LR, Calvo KR, Paul SM, Weir N, Brofferio A, Soldatos A, Biancotto A, Cowen EW, Digiovanna JJ, Gadina M, Lipton AJ, Hadigan C, Holland SM, Fontana J, Alawad AS, Brown RJ, Rother KI, Heller T, Brooks KM, Kumar P, Brooks SR, Waldman M, Singh HK, Nickeleit V, Silk M, Prakash A, Janes JM, Ozen S, Wakim PG, Brogan PA, Macias WL, Goldbach-Mansky R (2018) JAK1/2 inhibition with baricitinib in the treatment of autoinflammatory interferonopathies. J Clin Invest 128(7):3041–3052. https://​doi.​org/​10.​1172/​JCI98814CrossRefPubMedPubMedCentral
138.
Zurück zum Zitat Fremond ML, Rodero MP, Jeremiah N, Belot A, Jeziorski E, Duffy D, Bessis D, Cros G, Rice GI, Charbit B, Hulin A, Khoudour N, Caballero CM, Bodemer C, Fabre M, Berteloot L, Le Bourgeois M, Reix P, Walzer T, Moshous D, Blanche S, Fischer A, Bader-Meunier B, Rieux-Laucat F, Crow YJ, Neven B (2016) Efficacy of the Janus kinase 1/2 inhibitor ruxolitinib in the treatment of vasculopathy associated with TMEM173-activating mutations in 3 children. J Allergy Clin Immunol 138(6):1752–1755. https://doi.org/10.1016/j.jaci.2016.07.015CrossRefPubMed Fremond ML, Rodero MP, Jeremiah N, Belot A, Jeziorski E, Duffy D, Bessis D, Cros G, Rice GI, Charbit B, Hulin A, Khoudour N, Caballero CM, Bodemer C, Fabre M, Berteloot L, Le Bourgeois M, Reix P, Walzer T, Moshous D, Blanche S, Fischer A, Bader-Meunier B, Rieux-Laucat F, Crow YJ, Neven B (2016) Efficacy of the Janus kinase 1/2 inhibitor ruxolitinib in the treatment of vasculopathy associated with TMEM173-activating mutations in 3 children. J Allergy Clin Immunol 138(6):1752–1755. https://​doi.​org/​10.​1016/​j.​jaci.​2016.​07.​015CrossRefPubMed
139.
140.
Zurück zum Zitat Meesilpavikkai K, Dik WA, Schrijver B, van Helden-Meeuwsen CG, Versnel MA, van Hagen PM, Bijlsma EK, Ruivenkamp CAL, Oele MJ, Dalm V (2019) Efficacy of baricitinib in the treatment of chilblains associated with Aicardi-Goutieres syndrome, a type I interferonopathy. Arthritis Rheumatol (Hoboken, NJ) 71(5):829–831. https://doi.org/10.1002/art.40805CrossRef Meesilpavikkai K, Dik WA, Schrijver B, van Helden-Meeuwsen CG, Versnel MA, van Hagen PM, Bijlsma EK, Ruivenkamp CAL, Oele MJ, Dalm V (2019) Efficacy of baricitinib in the treatment of chilblains associated with Aicardi-Goutieres syndrome, a type I interferonopathy. Arthritis Rheumatol (Hoboken, NJ) 71(5):829–831. https://​doi.​org/​10.​1002/​art.​40805CrossRef
154.
Zurück zum Zitat Sarkar MK, Hile GA, Tsoi LC, Xing X, Liu J, Liang Y, Berthier CC, Swindell WR, Patrick MT, Shao S, Tsou PS, Uppala R, Beamer MA, Srivastava A, Bielas SL, Harms PW, Getsios S, Elder JT, Voorhees JJ, Gudjonsson JE, Kahlenberg JM (2018) Photosensitivity and type I IFN responses in cutaneous lupus are driven by epidermal-derived interferon kappa. Ann Rheum Dis 77(11):1653–1664. https://doi.org/10.1136/annrheumdis-2018-213197CrossRefPubMed Sarkar MK, Hile GA, Tsoi LC, Xing X, Liu J, Liang Y, Berthier CC, Swindell WR, Patrick MT, Shao S, Tsou PS, Uppala R, Beamer MA, Srivastava A, Bielas SL, Harms PW, Getsios S, Elder JT, Voorhees JJ, Gudjonsson JE, Kahlenberg JM (2018) Photosensitivity and type I IFN responses in cutaneous lupus are driven by epidermal-derived interferon kappa. Ann Rheum Dis 77(11):1653–1664. https://​doi.​org/​10.​1136/​annrheumdis-2018-213197CrossRefPubMed
157.
Zurück zum Zitat Wahadat MJ, Bodewes ILA, Maria NI, van Helden-Meeuwsen CG, van Dijk-Hummelman A, Steenwijk EC, Kamphuis S, Versnel MA (2018) Type I IFN signature in childhood-onset systemic lupus erythematosus: a conspiracy of DNA- and RNA-sensing receptors? Arthritis Res Therapy 20(1):4. https://doi.org/10.1186/s13075-017-1501-zCrossRef Wahadat MJ, Bodewes ILA, Maria NI, van Helden-Meeuwsen CG, van Dijk-Hummelman A, Steenwijk EC, Kamphuis S, Versnel MA (2018) Type I IFN signature in childhood-onset systemic lupus erythematosus: a conspiracy of DNA- and RNA-sensing receptors? Arthritis Res Therapy 20(1):4. https://​doi.​org/​10.​1186/​s13075-017-1501-zCrossRef
161.
Zurück zum Zitat McBride JM, Jiang J, Abbas AR, Morimoto A, Li J, Maciuca R, Townsend M, Wallace DJ, Kennedy WP, Drappa J (2012) Safety and pharmacodynamics of rontalizumab in patients with systemic lupus erythematosus: results of a phase I, placebo-controlled, double-blind, dose-escalation study. Arthritis Rheum 64(11):3666–3676. https://doi.org/10.1002/art.34632CrossRefPubMed McBride JM, Jiang J, Abbas AR, Morimoto A, Li J, Maciuca R, Townsend M, Wallace DJ, Kennedy WP, Drappa J (2012) Safety and pharmacodynamics of rontalizumab in patients with systemic lupus erythematosus: results of a phase I, placebo-controlled, double-blind, dose-escalation study. Arthritis Rheum 64(11):3666–3676. https://​doi.​org/​10.​1002/​art.​34632CrossRefPubMed
162.
Zurück zum Zitat Khamashta M, Merrill JT, Werth VP, Furie R, Kalunian K, Illei GG, Drappa J, Wang L, Greth W, investigators CDs (2016) Sifalimumab, an anti-interferon-alpha monoclonal antibody, in moderate to severe systemic lupus erythematosus: a randomised, double-blind, placebo-controlled study. Ann Rheum Dis 75 (11):1909–1916. https://doi.org/10.1136/annrheumdis-2015-208562 Khamashta M, Merrill JT, Werth VP, Furie R, Kalunian K, Illei GG, Drappa J, Wang L, Greth W, investigators CDs (2016) Sifalimumab, an anti-interferon-alpha monoclonal antibody, in moderate to severe systemic lupus erythematosus: a randomised, double-blind, placebo-controlled study. Ann Rheum Dis 75 (11):1909–1916. https://​doi.​org/​10.​1136/​annrheumdis-2015-208562
164.
Zurück zum Zitat Ducreux J, Houssiau FA, Vandepapeliere P, Jorgensen C, Lazaro E, Spertini F, Colaone F, Roucairol C, Laborie M, Croughs T, Grouard-Vogel G, Lauwerys BR (2016) Interferon alpha kinoid induces neutralizing anti-interferon alpha antibodies that decrease the expression of interferon-induced and B cell activation associated transcripts: analysis of extended follow-up data from the interferon alpha kinoid phase I/II study. Rheumatology (Oxford, England) 55(10):1901–1905. https://doi.org/10.1093/rheumatology/kew262CrossRef Ducreux J, Houssiau FA, Vandepapeliere P, Jorgensen C, Lazaro E, Spertini F, Colaone F, Roucairol C, Laborie M, Croughs T, Grouard-Vogel G, Lauwerys BR (2016) Interferon alpha kinoid induces neutralizing anti-interferon alpha antibodies that decrease the expression of interferon-induced and B cell activation associated transcripts: analysis of extended follow-up data from the interferon alpha kinoid phase I/II study. Rheumatology (Oxford, England) 55(10):1901–1905. https://​doi.​org/​10.​1093/​rheumatology/​kew262CrossRef
165.
166.
Zurück zum Zitat Furie R, Khamashta M, Merrill JT, Werth VP, Kalunian K, Brohawn P, Illei GG, Drappa J, Wang L, Yoo S, Investigators CDS (2017) Anifrolumab, an anti-interferon-alpha receptor monoclonal antibody, in moderate-to-severe systemic lupus erythematosus. Arthritis & rheumatology (Hoboken, NJ) 69(2):376–386. https://doi.org/10.1002/art.39962CrossRef Furie R, Khamashta M, Merrill JT, Werth VP, Kalunian K, Brohawn P, Illei GG, Drappa J, Wang L, Yoo S, Investigators CDS (2017) Anifrolumab, an anti-interferon-alpha receptor monoclonal antibody, in moderate-to-severe systemic lupus erythematosus. Arthritis & rheumatology (Hoboken, NJ) 69(2):376–386. https://​doi.​org/​10.​1002/​art.​39962CrossRef
169.
170.
Zurück zum Zitat Zhan Y, Carrington EM, Ko HJ, Vikstrom IB, Oon S, Zhang JG, Vremec D, Brady JL, Bouillet P, Wu L, Huang DC, Wicks IP, Morand EF, Strasser A, Lew AM (2015) Bcl-2 antagonists kill plasmacytoid dendritic cells from lupus-prone mice and dampen interferon-alpha production. Arthritis & rheumatology (Hoboken, NJ) 67(3):797–808. https://doi.org/10.1002/art.38966CrossRef Zhan Y, Carrington EM, Ko HJ, Vikstrom IB, Oon S, Zhang JG, Vremec D, Brady JL, Bouillet P, Wu L, Huang DC, Wicks IP, Morand EF, Strasser A, Lew AM (2015) Bcl-2 antagonists kill plasmacytoid dendritic cells from lupus-prone mice and dampen interferon-alpha production. Arthritis & rheumatology (Hoboken, NJ) 67(3):797–808. https://​doi.​org/​10.​1002/​art.​38966CrossRef
171.
Zurück zum Zitat Lu P, Fleischmann R, Curtis C, Ignatenko S, Clarke SH, Desai M, Wong SL, Grebe KM, Black K, Zeng J, Stolzenbach J, Medema JK (2018) Safety and pharmacodynamics of venetoclax (ABT-199) in a randomized single and multiple ascending dose study in women with systemic lupus erythematosus. Lupus 27(2):290–302. https://doi.org/10.1177/0961203317719334CrossRefPubMed Lu P, Fleischmann R, Curtis C, Ignatenko S, Clarke SH, Desai M, Wong SL, Grebe KM, Black K, Zeng J, Stolzenbach J, Medema JK (2018) Safety and pharmacodynamics of venetoclax (ABT-199) in a randomized single and multiple ascending dose study in women with systemic lupus erythematosus. Lupus 27(2):290–302. https://​doi.​org/​10.​1177/​0961203317719334​CrossRefPubMed
174.
Zurück zum Zitat Dudhgaonkar S, Ranade S, Nagar J, Subramani S, Prasad DS, Karunanithi P, Srivastava R, Venkatesh K, Selvam S, Krishnamurthy P, Mariappan TT, Saxena A, Fan L, Stetsko DK, Holloway DA, Li X, Zhu J, Yang WP, Ruepp S, Nair S, Santella J, Duncia J, Hynes J, McIntyre KW, Carman JA (2017) Selective IRAK4 inhibition attenuates disease in murine lupus models and demonstrates steroid sparing activity. J Immunol 198(3):1308–1319. https://doi.org/10.4049/jimmunol.1600583CrossRefPubMed Dudhgaonkar S, Ranade S, Nagar J, Subramani S, Prasad DS, Karunanithi P, Srivastava R, Venkatesh K, Selvam S, Krishnamurthy P, Mariappan TT, Saxena A, Fan L, Stetsko DK, Holloway DA, Li X, Zhu J, Yang WP, Ruepp S, Nair S, Santella J, Duncia J, Hynes J, McIntyre KW, Carman JA (2017) Selective IRAK4 inhibition attenuates disease in murine lupus models and demonstrates steroid sparing activity. J Immunol 198(3):1308–1319. https://​doi.​org/​10.​4049/​jimmunol.​1600583CrossRefPubMed
181.
Zurück zum Zitat Nader A, Stodtmann S, Friedel A, Mohamed MF, Othman AA (2020) Pharmacokinetics of Upadacitinib in healthy subjects and subjects with rheumatoid arthritis, Crohn’s disease, ulcerative colitis, or atopic dermatitis: population analyses of phase 1 and 2 clinical trials. J Clin Pharmacol 60(4):528–539. https://doi.org/10.1002/jcph.1550CrossRefPubMed Nader A, Stodtmann S, Friedel A, Mohamed MF, Othman AA (2020) Pharmacokinetics of Upadacitinib in healthy subjects and subjects with rheumatoid arthritis, Crohn’s disease, ulcerative colitis, or atopic dermatitis: population analyses of phase 1 and 2 clinical trials. J Clin Pharmacol 60(4):528–539. https://​doi.​org/​10.​1002/​jcph.​1550CrossRefPubMed
182.
Zurück zum Zitat Genovese MC, Kalunian K, Gottenberg JE, Mozaffarian N, Bartok B, Matzkies F, Gao J, Guo Y, Tasset C, Sundy JS, de Vlam K, Walker D, Takeuchi T (2019) Effect of filgotinib vs placebo on clinical response in patients with moderate to severe rheumatoid arthritis refractory to disease-modifying antirheumatic drug therapy: the FINCH 2 randomized clinical trial. JAMA 322(4):315–325. https://doi.org/10.1001/jama.2019.9055CrossRefPubMedPubMedCentral Genovese MC, Kalunian K, Gottenberg JE, Mozaffarian N, Bartok B, Matzkies F, Gao J, Guo Y, Tasset C, Sundy JS, de Vlam K, Walker D, Takeuchi T (2019) Effect of filgotinib vs placebo on clinical response in patients with moderate to severe rheumatoid arthritis refractory to disease-modifying antirheumatic drug therapy: the FINCH 2 randomized clinical trial. JAMA 322(4):315–325. https://​doi.​org/​10.​1001/​jama.​2019.​9055CrossRefPubMedPubMedCentral
183.
Zurück zum Zitat Takeuchi T, Tanaka Y, Tanaka S, Kawakami A, Iwasaki M, Katayama K, Rokuda M, Izutsu H, Ushijima S, Kaneko Y, Shiomi T, Yamada E, van der Heijde D (2019) Efficacy and safety of peficitinib (ASP015K) in patients with rheumatoid arthritis and an inadequate response to methotrexate: results of a phase III randomised, double-blind, placebo-controlled trial (RAJ4) in Japan. Ann Rheum Dis 78(10):1305–1319. https://doi.org/10.1136/annrheumdis-2019-215164CrossRefPubMed Takeuchi T, Tanaka Y, Tanaka S, Kawakami A, Iwasaki M, Katayama K, Rokuda M, Izutsu H, Ushijima S, Kaneko Y, Shiomi T, Yamada E, van der Heijde D (2019) Efficacy and safety of peficitinib (ASP015K) in patients with rheumatoid arthritis and an inadequate response to methotrexate: results of a phase III randomised, double-blind, placebo-controlled trial (RAJ4) in Japan. Ann Rheum Dis 78(10):1305–1319. https://​doi.​org/​10.​1136/​annrheumdis-2019-215164CrossRefPubMed
184.
Zurück zum Zitat Genovese MC, Greenwald M, Codding C, Zubrzycka-Sienkiewicz A, Kivitz AJ, Wang A, Shay K, Wang X, Garg JP, Cardiel MH (2017) Peficitinib, a JAK inhibitor, in combination with limited conventional synthetic disease-modifying antirheumatic drugs in the treatment of moderate-to-severe rheumatoid arthritis. Arthritis & rheumatology (Hoboken, NJ) 69(5):932–942. https://doi.org/10.1002/art.40054CrossRef Genovese MC, Greenwald M, Codding C, Zubrzycka-Sienkiewicz A, Kivitz AJ, Wang A, Shay K, Wang X, Garg JP, Cardiel MH (2017) Peficitinib, a JAK inhibitor, in combination with limited conventional synthetic disease-modifying antirheumatic drugs in the treatment of moderate-to-severe rheumatoid arthritis. Arthritis & rheumatology (Hoboken, NJ) 69(5):932–942. https://​doi.​org/​10.​1002/​art.​40054CrossRef
187.
Zurück zum Zitat Kjelgaard-Petersen CF, Platt A, Braddock M, Jenkins MA, Musa K, Graham E, Gantzel T, Slynn G, Weinblatt ME, Karsdal MA, Thudium CS, Bay-Jensen AC (2018) Translational biomarkers and ex vivo models of joint tissues as a tool for drug development in rheumatoid arthritis. Arthritis & rheumatology (Hoboken, NJ) 70(9):1419–1428. https://doi.org/10.1002/art.40527CrossRef Kjelgaard-Petersen CF, Platt A, Braddock M, Jenkins MA, Musa K, Graham E, Gantzel T, Slynn G, Weinblatt ME, Karsdal MA, Thudium CS, Bay-Jensen AC (2018) Translational biomarkers and ex vivo models of joint tissues as a tool for drug development in rheumatoid arthritis. Arthritis & rheumatology (Hoboken, NJ) 70(9):1419–1428. https://​doi.​org/​10.​1002/​art.​40527CrossRef
188.
Zurück zum Zitat Schafer PH, Kivitz AJ, Ma J, Korish S, Sutherland D, Li L, Azaryan A, Kosek J, Adams M, Capone L, Hur EM, Hough DR, Ringheim GE (2020) Spebrutinib (CC-292) affects markers of B cell activation, chemotaxis, and osteoclasts in patients with rheumatoid arthritis: results from a mechanistic study. Rheumatology and therapy 7(1):101–119. https://doi.org/10.1007/s40744-019-00182-7CrossRefPubMed Schafer PH, Kivitz AJ, Ma J, Korish S, Sutherland D, Li L, Azaryan A, Kosek J, Adams M, Capone L, Hur EM, Hough DR, Ringheim GE (2020) Spebrutinib (CC-292) affects markers of B cell activation, chemotaxis, and osteoclasts in patients with rheumatoid arthritis: results from a mechanistic study. Rheumatology and therapy 7(1):101–119. https://​doi.​org/​10.​1007/​s40744-019-00182-7CrossRefPubMed
190.
Zurück zum Zitat van Holten J, Pavelka K, Vencovsky J, Stahl H, Rozman B, Genovese M, Kivitz AJ, Alvaro J, Nuki G, Furst DE, Herrero-Beaumont G, McInnes IB, Musikic P, Tak PP (2005) A multicentre, randomised, double blind, placebo controlled phase II study of subcutaneous interferon beta-1a in the treatment of patients with active rheumatoid arthritis. Ann Rheum Dis 64(1):64–69. https://doi.org/10.1136/ard.2003.020347CrossRefPubMed van Holten J, Pavelka K, Vencovsky J, Stahl H, Rozman B, Genovese M, Kivitz AJ, Alvaro J, Nuki G, Furst DE, Herrero-Beaumont G, McInnes IB, Musikic P, Tak PP (2005) A multicentre, randomised, double blind, placebo controlled phase II study of subcutaneous interferon beta-1a in the treatment of patients with active rheumatoid arthritis. Ann Rheum Dis 64(1):64–69. https://​doi.​org/​10.​1136/​ard.​2003.​020347CrossRefPubMed
192.
Zurück zum Zitat Bracaglia C, de Graaf K, Pires Marafon D, Guilhot F, Ferlin W, Prencipe G, Caiello I, Davi S, Schulert G, Ravelli A, Grom AA, de Min C, De Benedetti F (2017) Elevated circulating levels of interferon-gamma and interferon-gamma-induced chemokines characterise patients with macrophage activation syndrome complicating systemic juvenile idiopathic arthritis. Ann Rheum Dis 76(1):166–172. https://doi.org/10.1136/annrheumdis-2015-209020CrossRefPubMed Bracaglia C, de Graaf K, Pires Marafon D, Guilhot F, Ferlin W, Prencipe G, Caiello I, Davi S, Schulert G, Ravelli A, Grom AA, de Min C, De Benedetti F (2017) Elevated circulating levels of interferon-gamma and interferon-gamma-induced chemokines characterise patients with macrophage activation syndrome complicating systemic juvenile idiopathic arthritis. Ann Rheum Dis 76(1):166–172. https://​doi.​org/​10.​1136/​annrheumdis-2015-209020CrossRefPubMed
193.
194.
196.
Zurück zum Zitat Ruperto N, Brunner HI, Zuber Z, Tzaribachev N, Kingsbury DJ, Foeldvari I, Horneff G, Smolewska E, Vehe RK, Hazra A, Wang R, Mebus CA, Alvey C, Lamba M, Krishnaswami S, Stock TC, Wang M, Suehiro R, Martini A, Lovell DJ, Pediatric Rheumatology International Trials O, Pediatric Rheumatology Collaborative Study G (2017) 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 Online J 15(1):86. https://doi.org/10.1186/s12969-017-0212-yCrossRefPubMedPubMedCentral Ruperto N, Brunner HI, Zuber Z, Tzaribachev N, Kingsbury DJ, Foeldvari I, Horneff G, Smolewska E, Vehe RK, Hazra A, Wang R, Mebus CA, Alvey C, Lamba M, Krishnaswami S, Stock TC, Wang M, Suehiro R, Martini A, Lovell DJ, Pediatric Rheumatology International Trials O, Pediatric Rheumatology Collaborative Study G (2017) 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 Online J 15(1):86. https://​doi.​org/​10.​1186/​s12969-017-0212-yCrossRefPubMedPubMedCentral
200.
Zurück zum Zitat Moneta GM, Pires Marafon D, Marasco E, Rosina S, Verardo M, Fiorillo C, Minetti C, Bracci-Laudiero L, Ravelli A, De Benedetti F, Nicolai R (2019) Muscle expression of type I and type II interferons is increased in juvenile dermatomyositis and related to clinical and histologic features. Arthritis & rheumatology (Hoboken, NJ) 71(6):1011–1021. https://doi.org/10.1002/art.40800CrossRef Moneta GM, Pires Marafon D, Marasco E, Rosina S, Verardo M, Fiorillo C, Minetti C, Bracci-Laudiero L, Ravelli A, De Benedetti F, Nicolai R (2019) Muscle expression of type I and type II interferons is increased in juvenile dermatomyositis and related to clinical and histologic features. Arthritis & rheumatology (Hoboken, NJ) 71(6):1011–1021. https://​doi.​org/​10.​1002/​art.​40800CrossRef
204.
Zurück zum Zitat Piper CJM, Wilkinson MGL, Deakin CT, Otto GW, Dowle S, Duurland CL, Adams S, Marasco E, Rosser EC, Radziszewska A, Carsetti R, Ioannou Y, Beales PL, Kelberman D, Isenberg DA, Mauri C, Nistala K, Wedderburn LR (2018) CD19(+)CD24(hi)CD38(hi) B cells are expanded in juvenile dermatomyositis and exhibit a pro-inflammatory phenotype after activation through toll-like receptor 7 and interferon-alpha. Front Immunol 9:1372. https://doi.org/10.3389/fimmu.2018.01372CrossRefPubMedPubMedCentral Piper CJM, Wilkinson MGL, Deakin CT, Otto GW, Dowle S, Duurland CL, Adams S, Marasco E, Rosser EC, Radziszewska A, Carsetti R, Ioannou Y, Beales PL, Kelberman D, Isenberg DA, Mauri C, Nistala K, Wedderburn LR (2018) CD19(+)CD24(hi)CD38(hi) B cells are expanded in juvenile dermatomyositis and exhibit a pro-inflammatory phenotype after activation through toll-like receptor 7 and interferon-alpha. Front Immunol 9:1372. https://​doi.​org/​10.​3389/​fimmu.​2018.​01372CrossRefPubMedPubMedCentral
205.
Zurück zum Zitat Gitiaux C, Latroche C, Weiss-Gayet M, Rodero MP, Duffy D, Bader-Meunier B, Glorion C, Nusbaum P, Bodemer C, Mouchiroud G, Chelly J, Germain S, Desguerre I, Chazaud B (2018) Myogenic progenitor cells exhibit type I interferon-driven proangiogenic properties and molecular signature during juvenile dermatomyositis. Arthritis & rheumatology (Hoboken, NJ) 70(1):134–145. https://doi.org/10.1002/art.40328CrossRef Gitiaux C, Latroche C, Weiss-Gayet M, Rodero MP, Duffy D, Bader-Meunier B, Glorion C, Nusbaum P, Bodemer C, Mouchiroud G, Chelly J, Germain S, Desguerre I, Chazaud B (2018) Myogenic progenitor cells exhibit type I interferon-driven proangiogenic properties and molecular signature during juvenile dermatomyositis. Arthritis & rheumatology (Hoboken, NJ) 70(1):134–145. https://​doi.​org/​10.​1002/​art.​40328CrossRef
206.
Zurück zum Zitat Ladislau L, Suarez-Calvet X, Toquet S, Landon-Cardinal O, Amelin D, Depp M, Rodero MP, Hathazi D, Duffy D, Bondet V, Preusse C, Bienvenu B, Rozenberg F, Roos A, Benjamim CF, Gallardo E, Illa I, Mouly V, Stenzel W, Butler-Browne G, Benveniste O, Allenbach Y (2018) JAK inhibitor improves type I interferon induced damage: proof of concept in dermatomyositis. Brain 141(6):1609–1621. https://doi.org/10.1093/brain/awy105CrossRefPubMed Ladislau L, Suarez-Calvet X, Toquet S, Landon-Cardinal O, Amelin D, Depp M, Rodero MP, Hathazi D, Duffy D, Bondet V, Preusse C, Bienvenu B, Rozenberg F, Roos A, Benjamim CF, Gallardo E, Illa I, Mouly V, Stenzel W, Butler-Browne G, Benveniste O, Allenbach Y (2018) JAK inhibitor improves type I interferon induced damage: proof of concept in dermatomyositis. Brain 141(6):1609–1621. https://​doi.​org/​10.​1093/​brain/​awy105CrossRefPubMed
209.
Zurück zum Zitat Higgs BW, Zhu W, Morehouse C, White WI, Brohawn P, Guo X, Rebelatto M, Le C, Amato A, Fiorentino D, Greenberg SA, Drappa J, Richman L, Greth W, Jallal B, Yao Y (2014) A phase 1b clinical trial evaluating sifalimumab, an anti-IFN-alpha monoclonal antibody, shows target neutralisation of a type I IFN signature in blood of dermatomyositis and polymyositis patients. Ann Rheum Dis 73(1):256–262. https://doi.org/10.1136/annrheumdis-2012-202794CrossRefPubMed Higgs BW, Zhu W, Morehouse C, White WI, Brohawn P, Guo X, Rebelatto M, Le C, Amato A, Fiorentino D, Greenberg SA, Drappa J, Richman L, Greth W, Jallal B, Yao Y (2014) A phase 1b clinical trial evaluating sifalimumab, an anti-IFN-alpha monoclonal antibody, shows target neutralisation of a type I IFN signature in blood of dermatomyositis and polymyositis patients. Ann Rheum Dis 73(1):256–262. https://​doi.​org/​10.​1136/​annrheumdis-2012-202794CrossRefPubMed
212.
Zurück zum Zitat Brkic Z, Maria NI, van Helden-Meeuwsen CG, van de Merwe JP, van Daele PL, Dalm VA, Wildenberg ME, Beumer W, Drexhage HA, Versnel MA (2013) Prevalence of interferon type I signature in CD14 monocytes of patients with Sjogren’s syndrome and association with disease activity and BAFF gene expression. Ann Rheum Dis 72(5):728–735. https://doi.org/10.1136/annrheumdis-2012-201381CrossRefPubMed Brkic Z, Maria NI, van Helden-Meeuwsen CG, van de Merwe JP, van Daele PL, Dalm VA, Wildenberg ME, Beumer W, Drexhage HA, Versnel MA (2013) Prevalence of interferon type I signature in CD14 monocytes of patients with Sjogren’s syndrome and association with disease activity and BAFF gene expression. Ann Rheum Dis 72(5):728–735. https://​doi.​org/​10.​1136/​annrheumdis-2012-201381CrossRefPubMed
213.
Zurück zum Zitat Gottenberg JE, Cagnard N, Lucchesi C, Letourneur F, Mistou S, Lazure T, Jacques S, Ba N, Ittah M, Lepajolec C, Labetoulle M, Ardizzone M, Sibilia J, Fournier C, Chiocchia G, Mariette X (2006) Activation of IFN pathways and plasmacytoid dendritic cell recruitment in target organs of primary Sjogren’s syndrome. Proc Natl Acad Sci U S A 103(8):2770–2775. https://doi.org/10.1073/pnas.0510837103CrossRefPubMedPubMedCentral Gottenberg JE, Cagnard N, Lucchesi C, Letourneur F, Mistou S, Lazure T, Jacques S, Ba N, Ittah M, Lepajolec C, Labetoulle M, Ardizzone M, Sibilia J, Fournier C, Chiocchia G, Mariette X (2006) Activation of IFN pathways and plasmacytoid dendritic cell recruitment in target organs of primary Sjogren’s syndrome. Proc Natl Acad Sci U S A 103(8):2770–2775. https://​doi.​org/​10.​1073/​pnas.​0510837103CrossRefPubMedPubMedCentral
215.
Zurück zum Zitat Lee J, Lee J, Kwok SK, Baek S, Jang SG, Hong SM, Min JW, Choi SS, Lee J, Cho ML, Park SH (2018) JAK-1 inhibition suppresses interferon-induced BAFF production in human salivary gland: potential therapeutic strategy for primary Sjogren’s syndrome. Arthritis & rheumatology (Hoboken, NJ) 70(12):2057–2066. https://doi.org/10.1002/art.40589CrossRef Lee J, Lee J, Kwok SK, Baek S, Jang SG, Hong SM, Min JW, Choi SS, Lee J, Cho ML, Park SH (2018) JAK-1 inhibition suppresses interferon-induced BAFF production in human salivary gland: potential therapeutic strategy for primary Sjogren’s syndrome. Arthritis & rheumatology (Hoboken, NJ) 70(12):2057–2066. https://​doi.​org/​10.​1002/​art.​40589CrossRef
217.
Zurück zum Zitat Bodewes ILA, Huijser E, van Helden-Meeuwsen CG, Tas L, Huizinga R, Dalm V, van Hagen PM, Groot N, Kamphuis S, van Daele PLA, Versnel MA (2018) TBK1: a key regulator and potential treatment target for interferon positive Sjogren’s syndrome, systemic lupus erythematosus and systemic sclerosis. J Autoimmun 91:97–102. https://doi.org/10.1016/j.jaut.2018.02.001CrossRefPubMed Bodewes ILA, Huijser E, van Helden-Meeuwsen CG, Tas L, Huizinga R, Dalm V, van Hagen PM, Groot N, Kamphuis S, van Daele PLA, Versnel MA (2018) TBK1: a key regulator and potential treatment target for interferon positive Sjogren’s syndrome, systemic lupus erythematosus and systemic sclerosis. J Autoimmun 91:97–102. https://​doi.​org/​10.​1016/​j.​jaut.​2018.​02.​001CrossRefPubMed
218.
224.
225.
Zurück zum Zitat Goldberg A, Geppert T, Schiopu E, Frech T, Hsu V, Simms RW, Peng SL, Yao Y, Elgeioushi N, Chang L, Wang B, Yoo S (2014) Dose-escalation of human anti-interferon-alpha receptor monoclonal antibody MEDI-546 in subjects with systemic sclerosis: a phase 1, multicenter, open label study. Arthritis research & therapy 16(1):R57. https://doi.org/10.1186/ar4492CrossRef Goldberg A, Geppert T, Schiopu E, Frech T, Hsu V, Simms RW, Peng SL, Yao Y, Elgeioushi N, Chang L, Wang B, Yoo S (2014) Dose-escalation of human anti-interferon-alpha receptor monoclonal antibody MEDI-546 in subjects with systemic sclerosis: a phase 1, multicenter, open label study. Arthritis research & therapy 16(1):R57. https://​doi.​org/​10.​1186/​ar4492CrossRef
Metadaten
Titel
Type I Interferons in the Pathogenesis and Treatment of Autoimmune Diseases
verfasst von
Jiao Jiang
Ming Zhao
Christopher Chang
Haijing Wu
Qianjin Lu
Publikationsdatum
17.06.2020
Verlag
Springer US
Erschienen in
Clinical Reviews in Allergy & Immunology / Ausgabe 2/2020
Print ISSN: 1080-0549
Elektronische ISSN: 1559-0267
DOI
https://doi.org/10.1007/s12016-020-08798-2

Weitere Artikel der Ausgabe 2/2020

Clinical Reviews in Allergy & Immunology 2/2020 Zur Ausgabe

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

HNO-Op. auch mit über 90?

16.04.2024 HNO-Chirurgie Nachrichten

Mit Blick auf das Risiko für Komplikationen nach elektiven Eingriffen im HNO-Bereich scheint das Alter der Patienten kein ausschlaggebender Faktor zu sein. Entscheidend ist offenbar, wie fit die Betroffenen tatsächlich sind.

Intrakapsuläre Tonsillektomie gewinnt an Boden

16.04.2024 Tonsillektomie Nachrichten

Gegenüber der vollständigen Entfernung der Gaumenmandeln hat die intrakapsuläre Tonsillektomie einige Vorteile, wie HNO-Fachleute aus den USA hervorheben. Sie haben die aktuelle Literatur zu dem Verfahren gesichtet.

Bilateraler Hörsturz hat eine schlechte Prognose

15.04.2024 Hörsturz Nachrichten

Die Mehrzahl der Menschen mit Hörsturz ist einseitig betroffen, doch auch ein beidseitiger Hörsturz ist möglich. Wie häufig solche Fälle sind und wie sich ihr Verlauf darstellt, hat eine HNO-Expertenrunde aus den USA untersucht.

Update HNO

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.