Skip to main content
Erschienen in: Clinical Reviews in Allergy & Immunology 2/2017

21.11.2016

An Update on the Use of Immunomodulators in Primary Immunodeficiencies

Erschienen in: Clinical Reviews in Allergy & Immunology | Ausgabe 2/2017

Einloggen, um Zugang zu erhalten

Abstract

The genomic revolution in the past decade fuelled by breathtaking advances in sequencing technologies has defined several new genetic diseases of the immune system. Many of these newly characterized diseases are a result of defects in genes involved in immune regulation. The discovery of these diseases has opened a vista of new therapeutic possibilities. Immunomodulatory agents, a hitherto unexplored therapeutic option in primary immunodeficiency diseases have been tried in a host of these newly described maladies. These agents have been shown conclusively to favorably modulate immune responses, resulting in abatement of clinical manifestations both in experimental models and patients. While some of the treatment options have been approved for therapeutic use or have been shown to be of merit in open-label trials, others have been shown to be efficacious in a handful of clinical cases, animal models, and cell lines. Interferon γ is approved for use in chronic granulomatous disease (CGD) to reduce the burden of infection and and has a good long-term efficacy. Recombinant human IL7 therapy has been shown increase the peripheral CD4 and CD8 T cell counts in patients with idiopathic CD4. Anti-IL1 agents are approved for the management of cryopyrin-related autoinflammatory syndrome, and their therapeutic efficacy is being increasingly recognized in other autoinflammatory syndromes and CGD. Mammalian target of rapamycin (mTOR) inhibitors have been proven useful in autoimmune lymphoproliferative syndrome (ALPS) and in IPEX syndrome. Therapies reported to be potential use in case reports include abatacept in CTLA4 haploinsufficiency and LRBA deficiency, ruxolitinib in gain-of-function STAT1, tocilizumab in gain-of-function STAT3 defect, mTOR inhibitors in PIK3CD activation, magnesium in XMEN syndrome, and pioglitazone in CGD. Treatment options of merit in human cell lines include interferon α and interferon β in TLR3 and UNC-93B deficiencies, anti-interferon therapy in SAVI, and Rho-kinase inhibitors in TTC7A deficiency. Anti-IL17 agents have show efficacy in animal models of leukocyte adhesion defect (LAD) and ALPS. This topical review explores the use of various immunomodulators and other biological agents in the context of primary immunodeficiency and autoinflammatory diseases.
Literatur
1.
Zurück zum Zitat Al-Herz W, Bousfiha A, Casanova J-L, Chatila T, Conley ME, Cunningham-Rundles C, Etzioni A, Franco JL, Gaspar HB, Holland SM, Klein C, Nonoyama S, Ochs HD, Oksenhendler E, Picard C, Puck JM, Sullivan K, Tang ML (2014) Primary immunodeficiency diseases: an update on the classification from the international union of immunological societies expert committee for primary immunodeficiency. Front Immunol 5:162. doi:10.3389/fimmu.2014.00162 PubMedPubMedCentral Al-Herz W, Bousfiha A, Casanova J-L, Chatila T, Conley ME, Cunningham-Rundles C, Etzioni A, Franco JL, Gaspar HB, Holland SM, Klein C, Nonoyama S, Ochs HD, Oksenhendler E, Picard C, Puck JM, Sullivan K, Tang ML (2014) Primary immunodeficiency diseases: an update on the classification from the international union of immunological societies expert committee for primary immunodeficiency. Front Immunol 5:162. doi:10.​3389/​fimmu.​2014.​00162 PubMedPubMedCentral
2.
Zurück zum Zitat Aguilar C, Malphettes M, Donadieu J, Chandesris O, Coignard-Biehler H, Catherinot E, Pellier I, Stephan JL et al (2014) Prevention of infections during primary immunodeficiency. Clin Infect Dis 59(10):1462–1470. doi:10.1093/cid/ciu646 PubMedCrossRef Aguilar C, Malphettes M, Donadieu J, Chandesris O, Coignard-Biehler H, Catherinot E, Pellier I, Stephan JL et al (2014) Prevention of infections during primary immunodeficiency. Clin Infect Dis 59(10):1462–1470. doi:10.​1093/​cid/​ciu646 PubMedCrossRef
4.
Zurück zum Zitat Griffith LM, Cowan MJ, Notarangelo LD, Puck JM, Buckley RH, Candotti F, Conley ME, Fleisher TA, Gaspar HB, Kohn DB, Ochs HD, O’Reilly RJ, Rizzo JD, Roifman CM, Small TN, Shearer WT (2009) Improving cellular therapy for primary immune deficiency diseases: recognition, diagnosis, and management. J Allergy Clin Immunol 124(6):1152–1160 . doi:10.1016/j.jaci.2009.10.022e12PubMedPubMedCentralCrossRef Griffith LM, Cowan MJ, Notarangelo LD, Puck JM, Buckley RH, Candotti F, Conley ME, Fleisher TA, Gaspar HB, Kohn DB, Ochs HD, O’Reilly RJ, Rizzo JD, Roifman CM, Small TN, Shearer WT (2009) Improving cellular therapy for primary immune deficiency diseases: recognition, diagnosis, and management. J Allergy Clin Immunol 124(6):1152–1160 . doi:10.​1016/​j.​jaci.​2009.​10.​022e12PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat ter Haar NM, Oswald M, Jeyaratnam J, Anton J, Barron KS, Brogan PA, Cantarini L, Galeotti C, Grateau G, Hentgen V, Hofer M, Kallinich T et al (2015) Recommendations for the management of autoinflammatory diseases. Ann Rheum Dis 74(9):1636–1644. doi:10.1136/annrheumdis-2015-207546 PubMedCrossRef ter Haar NM, Oswald M, Jeyaratnam J, Anton J, Barron KS, Brogan PA, Cantarini L, Galeotti C, Grateau G, Hentgen V, Hofer M, Kallinich T et al (2015) Recommendations for the management of autoinflammatory diseases. Ann Rheum Dis 74(9):1636–1644. doi:10.​1136/​annrheumdis-2015-207546 PubMedCrossRef
8.
Zurück zum Zitat Federici S, Sormani MP, Ozen S, Lachmann HJ, Amaryan G, Woo P, Koné-Paut I, Dewarrat N, Cantarini L, Insalaco A, Uziel Y, Rigante D (2015) Evidence-based provisional clinical classification criteria for autoinflammatory periodic fevers. Ann Rheum Dis 74(5):799–805. doi:10.1136/annrheumdis-2014-206580 PubMedCrossRef Federici S, Sormani MP, Ozen S, Lachmann HJ, Amaryan G, Woo P, Koné-Paut I, Dewarrat N, Cantarini L, Insalaco A, Uziel Y, Rigante D (2015) Evidence-based provisional clinical classification criteria for autoinflammatory periodic fevers. Ann Rheum Dis 74(5):799–805. doi:10.​1136/​annrheumdis-2014-206580 PubMedCrossRef
16.
Zurück zum Zitat Lafaille FG, Pessach IM, Zhang S-Y, Ciancanelli MJ, Herman M, Abhyankar A, Ying SW, Keros S, Goldstein PA, Mostoslavsky G, Ordovas-Montanes J, Jouanguy E et al (2012) Impaired intrinsic immunity to HSV-1 in human iPSC-derived TLR3-deficient CNS cells. Nature 491(7426):769–773. doi:10.1038/nature11583 PubMedPubMedCentral Lafaille FG, Pessach IM, Zhang S-Y, Ciancanelli MJ, Herman M, Abhyankar A, Ying SW, Keros S, Goldstein PA, Mostoslavsky G, Ordovas-Montanes J, Jouanguy E et al (2012) Impaired intrinsic immunity to HSV-1 in human iPSC-derived TLR3-deficient CNS cells. Nature 491(7426):769–773. doi:10.​1038/​nature11583 PubMedPubMedCentral
17.
Zurück zum Zitat The International Chronic Granulomatous Disease Cooperative Study Group (1991) A controlled trial of interferon gamma to prevent infection in chronic granulomatous disease. N Engl J Med 324(8):509–516. doi:10.1056/NEJM19910221324080 CrossRef The International Chronic Granulomatous Disease Cooperative Study Group (1991) A controlled trial of interferon gamma to prevent infection in chronic granulomatous disease. N Engl J Med 324(8):509–516. doi:10.​1056/​NEJM199102213240​80 CrossRef
18.
Zurück zum Zitat Weening RS, Leitz GJ, Seger RA (1995) Recombinant human interferon-gamma in patients with chronic granulomatous disease—European follow up study. Eur J Pediatr 154(4):295–298. doi:10.1007/BF01957365 PubMed Weening RS, Leitz GJ, Seger RA (1995) Recombinant human interferon-gamma in patients with chronic granulomatous disease—European follow up study. Eur J Pediatr 154(4):295–298. doi:10.​1007/​BF01957365 PubMed
20.
Zurück zum Zitat Bemiller LS, Roberts DH, Starko KM, Curnutte JT (1995) Safety and effectiveness of long-term interferon gamma therapy in patients with chronic granulomatous disease. Blood Cells Mol Dis 21(3):239–247. doi:10.1006/bcmd.1995.0028 PubMedCrossRef Bemiller LS, Roberts DH, Starko KM, Curnutte JT (1995) Safety and effectiveness of long-term interferon gamma therapy in patients with chronic granulomatous disease. Blood Cells Mol Dis 21(3):239–247. doi:10.​1006/​bcmd.​1995.​0028 PubMedCrossRef
21.
Zurück zum Zitat Marciano BE, Wesley R, De Carlo ES, Anderson VL, Barnhart LA, Darnell D, Malech HL, Gallin JI, Holland SM (2004) Long-term interferon-gamma therapy for patients with chronic granulomatous disease. Clin Infect Dis 39(5):692–699. doi:10.1086/422993 PubMedCrossRef Marciano BE, Wesley R, De Carlo ES, Anderson VL, Barnhart LA, Darnell D, Malech HL, Gallin JI, Holland SM (2004) Long-term interferon-gamma therapy for patients with chronic granulomatous disease. Clin Infect Dis 39(5):692–699. doi:10.​1086/​422993 PubMedCrossRef
22.
Zurück zum Zitat Ahlin A, Elinder G, Palmblad J (1997) Dose-dependent enhancements by interferon-gamma on functional responses of neutrophils from chronic granulomatous disease patients. Blood 89(9):3396–3401PubMed Ahlin A, Elinder G, Palmblad J (1997) Dose-dependent enhancements by interferon-gamma on functional responses of neutrophils from chronic granulomatous disease patients. Blood 89(9):3396–3401PubMed
23.
Zurück zum Zitat Naderi beni F, Fattahi F, Mirshafiey A, Ansari M, Mohsenzadegan M, Movahedi M, Pourpak Z, Moin M (2012) Increased production of nitric oxide by neutrophils from patients with chronic granulomatous disease on interferon-gamma treatment. Int Immunopharmacol 12(4):689–693. doi:10.1016/j.intimp.2012.01.016 PubMedCrossRef Naderi beni F, Fattahi F, Mirshafiey A, Ansari M, Mohsenzadegan M, Movahedi M, Pourpak Z, Moin M (2012) Increased production of nitric oxide by neutrophils from patients with chronic granulomatous disease on interferon-gamma treatment. Int Immunopharmacol 12(4):689–693. doi:10.​1016/​j.​intimp.​2012.​01.​016 PubMedCrossRef
25.
28.
Zurück zum Zitat Sharma VK, Pai G, Deswarte C, Lodha R, Singh S, Kang LW, Yin CC, Casanova JL, Bustamante J, Kabra SK (2015) Disseminated Mycobacterium avium complex infection in a child with partial dominant interferon gamma receptor 1 deficiency in India. J Clin Immunol 35(5):459–462. doi:10.1007/s10875-015-0173-1 PubMedCrossRef Sharma VK, Pai G, Deswarte C, Lodha R, Singh S, Kang LW, Yin CC, Casanova JL, Bustamante J, Kabra SK (2015) Disseminated Mycobacterium avium complex infection in a child with partial dominant interferon gamma receptor 1 deficiency in India. J Clin Immunol 35(5):459–462. doi:10.​1007/​s10875-015-0173-1 PubMedCrossRef
29.
Zurück zum Zitat Takeda K, Kawai T, Nakazawa Y, Komuro H, Shoji K, Morita K, Katsuta T, Yamamoto M, Miyairi I, Ohya Y, Ishiguro A, Onodera M (2014) Augmentation of antitubercular therapy with IFNγ in a patient with dominant partial IFNγ receptor 1 deficiency. Clin Immunol 151(1):25–28. doi:10.1016/j.clim.2014.01.004 PubMedCrossRef Takeda K, Kawai T, Nakazawa Y, Komuro H, Shoji K, Morita K, Katsuta T, Yamamoto M, Miyairi I, Ohya Y, Ishiguro A, Onodera M (2014) Augmentation of antitubercular therapy with IFNγ in a patient with dominant partial IFNγ receptor 1 deficiency. Clin Immunol 151(1):25–28. doi:10.​1016/​j.​clim.​2014.​01.​004 PubMedCrossRef
30.
Zurück zum Zitat Alangari AA, Al-Zamil F, Al-Mazrou A, Al-Muhsen S, Boisson-Dupuis S, Awadallah S, Kambal A, Casanova JL (2011) Treatment of disseminated mycobacterial infection with high-dose IFN-γ in a patient with IL-12Rβ1 deficiency. Clin Dev Immunol 2011:691956. doi:10.1155/2011/691956 PubMedCrossRef Alangari AA, Al-Zamil F, Al-Mazrou A, Al-Muhsen S, Boisson-Dupuis S, Awadallah S, Kambal A, Casanova JL (2011) Treatment of disseminated mycobacterial infection with high-dose IFN-γ in a patient with IL-12Rβ1 deficiency. Clin Dev Immunol 2011:691956. doi:10.​1155/​2011/​691956 PubMedCrossRef
31.
32.
Zurück zum Zitat Azuma H, Sakata H, Saijyou M, Okuno A (1993) Effect of interleukin 2 on intractable herpes virus infection and chronic eczematoid dermatitis in a patient with Wiskott-Aldrich syndrome. Eur J Pediatr 152(12):998–1000PubMedCrossRef Azuma H, Sakata H, Saijyou M, Okuno A (1993) Effect of interleukin 2 on intractable herpes virus infection and chronic eczematoid dermatitis in a patient with Wiskott-Aldrich syndrome. Eur J Pediatr 152(12):998–1000PubMedCrossRef
33.
Zurück zum Zitat Azuma H, Oshima M, Ito K, Okuno A, Kawabata I, Banba K, Murahashi H, Sekine T, Kato Y, Ikebuchi K, Ikeda H (2000) Impaired interleukin-2 production in T-cells from a patient with Wiskott-Aldrich syndrome: basis of clinical effect of interleukin-2 replacement therapy. Eur J Pediatr 159(8):633–634PubMedCrossRef Azuma H, Oshima M, Ito K, Okuno A, Kawabata I, Banba K, Murahashi H, Sekine T, Kato Y, Ikebuchi K, Ikeda H (2000) Impaired interleukin-2 production in T-cells from a patient with Wiskott-Aldrich syndrome: basis of clinical effect of interleukin-2 replacement therapy. Eur J Pediatr 159(8):633–634PubMedCrossRef
34.
Zurück zum Zitat Orange JS, Roy-Ghanta S, Mace EM, Maru S, Rak GD, Sanborn KB, Fasth A, Saltzman R, Paisley A, Monaco-Shawver L, Banerjee PP, Pandey R (2011) IL-2 induces a WAVE2-dependent pathway for actin reorganization that enables WASp-independent human NK cell function. J Clin Invest 121(4):1535–1548. doi:10.1172/JCI44862 PubMedPubMedCentralCrossRef Orange JS, Roy-Ghanta S, Mace EM, Maru S, Rak GD, Sanborn KB, Fasth A, Saltzman R, Paisley A, Monaco-Shawver L, Banerjee PP, Pandey R (2011) IL-2 induces a WAVE2-dependent pathway for actin reorganization that enables WASp-independent human NK cell function. J Clin Invest 121(4):1535–1548. doi:10.​1172/​JCI44862 PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Myrup B, Valerius NH, Mortensen PB (1998) Treatment of enteritis in chronic granulomatous disease with granulocyte colony stimulating factor. Gut 42(1):127–130PubMedPubMedCentralCrossRef Myrup B, Valerius NH, Mortensen PB (1998) Treatment of enteritis in chronic granulomatous disease with granulocyte colony stimulating factor. Gut 42(1):127–130PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Aksentijevich I, Masters SL, Ferguson PJ, Dancey P, Frenkel J, van Royen-Kerkhoff A, Laxer R, Tedgard U, Cowen EW, Pham TH, Booty M, Estes JD et al (2009) An autoinflammatory disease with deficiency of the interleukin-1-receptor antagonist. N Engl J Med 360(23):2426–2437. doi:10.1056/NEJMoa0807865 PubMedPubMedCentralCrossRef Aksentijevich I, Masters SL, Ferguson PJ, Dancey P, Frenkel J, van Royen-Kerkhoff A, Laxer R, Tedgard U, Cowen EW, Pham TH, Booty M, Estes JD et al (2009) An autoinflammatory disease with deficiency of the interleukin-1-receptor antagonist. N Engl J Med 360(23):2426–2437. doi:10.​1056/​NEJMoa0807865 PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Cohen SB, Moreland LW, Cush JJ, Greenwald MW, Block S, Shergy WJ, Hanrahan PS, Kraishi MM, Patel A, Sun G, Bear MB (2004) A multicentre, double blind, randomised, placebo controlled trial of anakinra (Kineret), a recombinant interleukin 1 receptor antagonist, in patients with rheumatoid arthritis treated with background methotrexate. Ann Rheum Dis 63(9):1062–1068. doi:10.1136/ard.2003.016014 PubMedPubMedCentralCrossRef Cohen SB, Moreland LW, Cush JJ, Greenwald MW, Block S, Shergy WJ, Hanrahan PS, Kraishi MM, Patel A, Sun G, Bear MB (2004) A multicentre, double blind, randomised, placebo controlled trial of anakinra (Kineret), a recombinant interleukin 1 receptor antagonist, in patients with rheumatoid arthritis treated with background methotrexate. Ann Rheum Dis 63(9):1062–1068. doi:10.​1136/​ard.​2003.​016014 PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Hoffman HM, Throne ML, Amar NJ, Cartwright RC, Kivitz AJ, Soo Y, Weinstein SP (2012) Long-term efficacy and safety profile of rilonacept in the treatment of cryopryin-associated periodic syndromes: results of a 72-week open-label extension study. Clin Ther 34(10):2091–2103. doi:10.1016/j.clinthera.2012.09.009 PubMedCrossRef Hoffman HM, Throne ML, Amar NJ, Cartwright RC, Kivitz AJ, Soo Y, Weinstein SP (2012) Long-term efficacy and safety profile of rilonacept in the treatment of cryopryin-associated periodic syndromes: results of a 72-week open-label extension study. Clin Ther 34(10):2091–2103. doi:10.​1016/​j.​clinthera.​2012.​09.​009 PubMedCrossRef
42.
Zurück zum Zitat Goldbach-Mansky R, Shroff SD, Wilson M, Snyder C, Plehn S, Barham B, Pham TH, Pucino F, Wesley RA, Papadopaulos JH, Weinstein SP, Mellis SJ, Kastner DL (2008) A pilot study to evaluate the safety and efficacy of the long-acting interleukin-1 inhibitor rilonacept (interleukin-1 trap) in patients with familial cold autoinflammatory syndrome. Arthritis Rheum 58(8):2432–2442. doi:10.1002/art.23620 PubMedPubMedCentralCrossRef Goldbach-Mansky R, Shroff SD, Wilson M, Snyder C, Plehn S, Barham B, Pham TH, Pucino F, Wesley RA, Papadopaulos JH, Weinstein SP, Mellis SJ, Kastner DL (2008) A pilot study to evaluate the safety and efficacy of the long-acting interleukin-1 inhibitor rilonacept (interleukin-1 trap) in patients with familial cold autoinflammatory syndrome. Arthritis Rheum 58(8):2432–2442. doi:10.​1002/​art.​23620 PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Lachmann HJ, Kone-Paut I, Kuemmerle-Deschner JB, Leslie KS, Hachulla E, Quartier P, Gitton X, Widmer A, Patel N, Hawkins PN (2009) Use of canakinumab in the cryopyrin-associated periodic syndrome. N Engl J Med 360(23):2416–2425. doi:10.1056/NEJMoa0810787 PubMedCrossRef Lachmann HJ, Kone-Paut I, Kuemmerle-Deschner JB, Leslie KS, Hachulla E, Quartier P, Gitton X, Widmer A, Patel N, Hawkins PN (2009) Use of canakinumab in the cryopyrin-associated periodic syndrome. N Engl J Med 360(23):2416–2425. doi:10.​1056/​NEJMoa0810787 PubMedCrossRef
44.
Zurück zum Zitat de Luca A, Smeekens SP, Casagrande A, Iannitti R, Conway KL, Gresnigt MS, Begun J, Plantinga TS, Joosten LA, van der Meer JW, Chamilos G, Netea MG et al (2014) IL-1 receptor blockade restores autophagy and reduces inflammation in chronic granulomatous disease in mice and in humans. Proc Natl Acad Sci U S A 111(9):3526–3531. doi:10.1073/pnas.1322831111 PubMedPubMedCentralCrossRef de Luca A, Smeekens SP, Casagrande A, Iannitti R, Conway KL, Gresnigt MS, Begun J, Plantinga TS, Joosten LA, van der Meer JW, Chamilos G, Netea MG et al (2014) IL-1 receptor blockade restores autophagy and reduces inflammation in chronic granulomatous disease in mice and in humans. Proc Natl Acad Sci U S A 111(9):3526–3531. doi:10.​1073/​pnas.​1322831111 PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat van de Veerdonk FL, Netea MG, Dinarello CA, van der Meer JWM (2011) Anakinra for the inflammatory complications of chronic granulomatous disease. Neth J Med 69(2):95PubMed van de Veerdonk FL, Netea MG, Dinarello CA, van der Meer JWM (2011) Anakinra for the inflammatory complications of chronic granulomatous disease. Neth J Med 69(2):95PubMed
46.
Zurück zum Zitat Schubert D, Bode C, Kenefeck R, Hou TZ, Wing JB, Kennedy A, Bulashevska A, Petersen BS, Schaffer AA, Gruning BA, Unger S, Frede N et al (2014) Autosomal dominant immune dysregulation syndrome in humans with CTLA4 mutations. Nat Med 20(12):1410–1416. doi:10.1038/nm.3746 PubMedPubMedCentralCrossRef Schubert D, Bode C, Kenefeck R, Hou TZ, Wing JB, Kennedy A, Bulashevska A, Petersen BS, Schaffer AA, Gruning BA, Unger S, Frede N et al (2014) Autosomal dominant immune dysregulation syndrome in humans with CTLA4 mutations. Nat Med 20(12):1410–1416. doi:10.​1038/​nm.​3746 PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Lee S, Moon JS, Lee C-R, Kim H-E, Baek S-M, Hwang S, Kang GH, Seo JK, Shin CH, Kang HJ, Ko JS, Park SG, Choi M (2016) Abatacept alleviates severe autoimmune symptoms in a patient carrying a de novo variant in CTLA-4. J Allergy Clin Immunol 137(1):327–330. doi:10.1016/j.jaci.2015.08.036 PubMedCrossRef Lee S, Moon JS, Lee C-R, Kim H-E, Baek S-M, Hwang S, Kang GH, Seo JK, Shin CH, Kang HJ, Ko JS, Park SG, Choi M (2016) Abatacept alleviates severe autoimmune symptoms in a patient carrying a de novo variant in CTLA-4. J Allergy Clin Immunol 137(1):327–330. doi:10.​1016/​j.​jaci.​2015.​08.​036 PubMedCrossRef
48.
Zurück zum Zitat Lo B, Zhang K, Lu W, Zheng L, Zhang Q, Kanellopoulou C, Zhang Y, Liu Z, Fritz JM, Marsh R, Husami A, Kissell D et al (2015) AUTOIMMUNE DISEASE. Patients with LRBA deficiency show CTLA4 loss and immune dysregulation responsive to abatacept therapy. Science 349(6246):436–440. doi:10.1126/science.aaa1663 PubMedCrossRef Lo B, Zhang K, Lu W, Zheng L, Zhang Q, Kanellopoulou C, Zhang Y, Liu Z, Fritz JM, Marsh R, Husami A, Kissell D et al (2015) AUTOIMMUNE DISEASE. Patients with LRBA deficiency show CTLA4 loss and immune dysregulation responsive to abatacept therapy. Science 349(6246):436–440. doi:10.​1126/​science.​aaa1663 PubMedCrossRef
49.
Zurück zum Zitat Alkhairy OK, Abolhassani H, Rezaei N, Fang M, Andersen KK, Chavoshzadeh Z, Mohammadzadeh I, El-Rajab MA, Massaad M, Chou J, Aghamohammadi A, Geha RS et al (2016) Spectrum of phenotypes associated with mutations in LRBA. J Clin Immunol 36(1):33–45. doi:10.1007/s10875-015-0224-7 PubMedCrossRef Alkhairy OK, Abolhassani H, Rezaei N, Fang M, Andersen KK, Chavoshzadeh Z, Mohammadzadeh I, El-Rajab MA, Massaad M, Chou J, Aghamohammadi A, Geha RS et al (2016) Spectrum of phenotypes associated with mutations in LRBA. J Clin Immunol 36(1):33–45. doi:10.​1007/​s10875-015-0224-7 PubMedCrossRef
51.
Zurück zum Zitat Moutsopoulos NM, Konkel J, Sarmadi M, Eskan MA, Wild T, Dutzan N, Abusleme L, Zenobia C, Hosur KB, Abe T, Uzel G, Chen W et al (2014) Defective neutrophil recruitment in leukocyte adhesion deficiency type I disease causes local IL-17-driven inflammatory bone loss. Sci Transl Med 6(229):229ra40. doi:10.1126/scitranslmed.3007696 PubMedPubMedCentralCrossRef Moutsopoulos NM, Konkel J, Sarmadi M, Eskan MA, Wild T, Dutzan N, Abusleme L, Zenobia C, Hosur KB, Abe T, Uzel G, Chen W et al (2014) Defective neutrophil recruitment in leukocyte adhesion deficiency type I disease causes local IL-17-driven inflammatory bone loss. Sci Transl Med 6(229):229ra40. doi:10.​1126/​scitranslmed.​3007696 PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Milner JD, Vogel TP, Forbes L, Ma CA, Stray-Pedersen A, Niemela JE, Lyons JJ, Engelhardt KR, Zhang Y, Topcagic N, Roberson ED, Matthews H et al (2015) Early-onset lymphoproliferation and autoimmunity caused by germline STAT3 gain-of-function mutations. Blood 125(4):591–599. doi:10.1182/blood-2014-09-602763 PubMedPubMedCentralCrossRef Milner JD, Vogel TP, Forbes L, Ma CA, Stray-Pedersen A, Niemela JE, Lyons JJ, Engelhardt KR, Zhang Y, Topcagic N, Roberson ED, Matthews H et al (2015) Early-onset lymphoproliferation and autoimmunity caused by germline STAT3 gain-of-function mutations. Blood 125(4):591–599. doi:10.​1182/​blood-2014-09-602763 PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Put K, Avau A, Brisse E, Mitera T, Put S, Proost P, Bader-Meunier B, Westhovens R, Van den Eynde BJ, Orabona S, Fallarino F, De Somer L et al (2015) Cytokines in systemic juvenile idiopathic arthritis and haemophagocytic lymphohistiocytosis: tipping the balance between interleukin-18 and interferon-γ. Rheumatology (Oxford) 54(8):1507–1517. doi:10.1093/rheumatology/keu524 CrossRef Put K, Avau A, Brisse E, Mitera T, Put S, Proost P, Bader-Meunier B, Westhovens R, Van den Eynde BJ, Orabona S, Fallarino F, De Somer L et al (2015) Cytokines in systemic juvenile idiopathic arthritis and haemophagocytic lymphohistiocytosis: tipping the balance between interleukin-18 and interferon-γ. Rheumatology (Oxford) 54(8):1507–1517. doi:10.​1093/​rheumatology/​keu524 CrossRef
57.
58.
Zurück zum Zitat Bracaglia C, Gatto A, Pardeo M, Lapeyre G, Ferlin W, Nelson R, de Min C, De Benedetti F (2015) Anti interferon-gamma (IFNγ) monoclonal antibody treatment in a patient carrying an NLRC4 mutation and severe hemophagocytic lymphohistiocytosis. Pediatr Rheumatol Online J 13(Suppl 1):O68. doi:10.1186/1546-0096-13-S1-O68 PubMedCentralCrossRef Bracaglia C, Gatto A, Pardeo M, Lapeyre G, Ferlin W, Nelson R, de Min C, De Benedetti F (2015) Anti interferon-gamma (IFNγ) monoclonal antibody treatment in a patient carrying an NLRC4 mutation and severe hemophagocytic lymphohistiocytosis. Pediatr Rheumatol Online J 13(Suppl 1):O68. doi:10.​1186/​1546-0096-13-S1-O68 PubMedCentralCrossRef
59.
Zurück zum Zitat Bulua AC, Mogul DB, Aksentijevich I, Singh H, He DY, Muenz LR, Ward MM, Yarboro CH, Kastner DL, Siegel RM, Hull KM (2012) Efficacy of etanercept in the tumor necrosis factor receptor-associated periodic syndrome: a prospective, open-label, dose-escalation study. Arthritis Rheum 64(3):908–913. doi:10.1002/art.33416 PubMedCrossRef Bulua AC, Mogul DB, Aksentijevich I, Singh H, He DY, Muenz LR, Ward MM, Yarboro CH, Kastner DL, Siegel RM, Hull KM (2012) Efficacy of etanercept in the tumor necrosis factor receptor-associated periodic syndrome: a prospective, open-label, dose-escalation study. Arthritis Rheum 64(3):908–913. doi:10.​1002/​art.​33416 PubMedCrossRef
61.
Zurück zum Zitat Marks DJB, Miyagi K, Rahman FZ, Novelli M, Bloom SL, Segal AW (2009) Inflammatory bowel disease in CGD reproduces the clinicopathological features of Crohn’s disease. Am J Gastroenterol 104(1):117–124. doi:10.1038/ajg.2008.72 PubMedCrossRef Marks DJB, Miyagi K, Rahman FZ, Novelli M, Bloom SL, Segal AW (2009) Inflammatory bowel disease in CGD reproduces the clinicopathological features of Crohn’s disease. Am J Gastroenterol 104(1):117–124. doi:10.​1038/​ajg.​2008.​72 PubMedCrossRef
62.
63.
Zurück zum Zitat Chase NM, Verbsky JW, Hintermeyer MK, Waukau JK, Tomita-Mitchell A, Casper JT, Singh S, Shahir KS, Tisol WB, Nugent ML, Rao RN, Mackinnon AC et al (2013) Use of combination chemotherapy for treatment of granulomatous and lymphocytic interstitial lung disease (GLILD) in patients with common variable immunodeficiency (CVID). J Clin Immunol 33(1):30–39. doi:10.1007/s10875-012-9755-3 PubMedCrossRef Chase NM, Verbsky JW, Hintermeyer MK, Waukau JK, Tomita-Mitchell A, Casper JT, Singh S, Shahir KS, Tisol WB, Nugent ML, Rao RN, Mackinnon AC et al (2013) Use of combination chemotherapy for treatment of granulomatous and lymphocytic interstitial lung disease (GLILD) in patients with common variable immunodeficiency (CVID). J Clin Immunol 33(1):30–39. doi:10.​1007/​s10875-012-9755-3 PubMedCrossRef
64.
Zurück zum Zitat Boursiquot J-N, Gérard L, Malphettes M, Fieschi C, Galicier L, Boutboul D, Borie R, Viallard JF, Soulas-Sprauel P, Berezne A, Jaccard A, Hachulla E et al (2013) Granulomatous disease in CVID: retrospective analysis of clinical characteristics and treatment efficacy in a cohort of 59 patients. J Clin Immunol 33(1):84–95. doi:10.1007/s10875-012-9778-9 PubMedCrossRef Boursiquot J-N, Gérard L, Malphettes M, Fieschi C, Galicier L, Boutboul D, Borie R, Viallard JF, Soulas-Sprauel P, Berezne A, Jaccard A, Hachulla E et al (2013) Granulomatous disease in CVID: retrospective analysis of clinical characteristics and treatment efficacy in a cohort of 59 patients. J Clin Immunol 33(1):84–95. doi:10.​1007/​s10875-012-9778-9 PubMedCrossRef
65.
Zurück zum Zitat Gobert D, Bussel JB, Cunningham-Rundles C, Galicier L, Dechartres A, Berezne A, Bonnotte B, DeRevel T, Auzary C, Jaussaud R, Larroche C, LeQuellec A et al (2011) Efficacy and safety of rituximab in common variable immunodeficiency-associated immune cytopenias: a retrospective multicentre study on 33 patients. Br J Haematol 155(4):498–508. doi:10.1111/j.1365-2141.2011.08880.x PubMedPubMedCentralCrossRef Gobert D, Bussel JB, Cunningham-Rundles C, Galicier L, Dechartres A, Berezne A, Bonnotte B, DeRevel T, Auzary C, Jaussaud R, Larroche C, LeQuellec A et al (2011) Efficacy and safety of rituximab in common variable immunodeficiency-associated immune cytopenias: a retrospective multicentre study on 33 patients. Br J Haematol 155(4):498–508. doi:10.​1111/​j.​1365-2141.​2011.​08880.​x PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Toyoda H, Azuma E, Kawasaki Y, Iwasa T, Ohashi H, Otsuki S, Iwamoto S, Hirayama M, Itoh-Habe N, Wada H, Kondo M, Keida Y, Ito T, Komada Y (2013) Cord blood transplantation combined with rituximab for Wiskott-Aldrich syndrome with autoimmune thrombotic thrombocytopenic purpura. J Allergy Clin Immunol 132(1):226–227. doi:10.1016/j.jaci.2013.01.040 PubMedCrossRef Toyoda H, Azuma E, Kawasaki Y, Iwasa T, Ohashi H, Otsuki S, Iwamoto S, Hirayama M, Itoh-Habe N, Wada H, Kondo M, Keida Y, Ito T, Komada Y (2013) Cord blood transplantation combined with rituximab for Wiskott-Aldrich syndrome with autoimmune thrombotic thrombocytopenic purpura. J Allergy Clin Immunol 132(1):226–227. doi:10.​1016/​j.​jaci.​2013.​01.​040 PubMedCrossRef
68.
Zurück zum Zitat Emir S, Vezir E, Azkur D, Demir HA, Metin A (2013) Characteristics of children with non-hodgkin lymphoma associated with primary immune deficiency diseases: descriptions of five patients. Pediatr Hematol Oncol 30(6):544–553. doi:10.3109/08880018.2013.792893 PubMedCrossRef Emir S, Vezir E, Azkur D, Demir HA, Metin A (2013) Characteristics of children with non-hodgkin lymphoma associated with primary immune deficiency diseases: descriptions of five patients. Pediatr Hematol Oncol 30(6):544–553. doi:10.​3109/​08880018.​2013.​792893 PubMedCrossRef
69.
Zurück zum Zitat Sebire NJ, Haselden S, Malone M, Davies EG, Ramsay AD (2003) Isolated EBV lymphoproliferative disease in a child with Wiskott-Aldrich syndrome manifesting as cutaneous lymphomatoid granulomatosis and responsive to anti-CD20 immunotherapy. J Clin Pathol 56(7):555–557. doi:10.1136/jcp.56.7.555 PubMedPubMedCentralCrossRef Sebire NJ, Haselden S, Malone M, Davies EG, Ramsay AD (2003) Isolated EBV lymphoproliferative disease in a child with Wiskott-Aldrich syndrome manifesting as cutaneous lymphomatoid granulomatosis and responsive to anti-CD20 immunotherapy. J Clin Pathol 56(7):555–557. doi:10.​1136/​jcp.​56.​7.​555 PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Browne SK, Zaman R, Sampaio EP, Jutivorakool K, Rosen LB, Ding L, Pancholi MJ, Yang LM, Priel DL, Uzel G, Freeman AF, Hayes CE et al (2012) Anti-CD20 (rituximab) therapy for anti–IFN-γ autoantibody–associated nontuberculous mycobacterial infection. Blood 119(17):3933–3939. doi:10.1182/blood-2011-12-395707 PubMedPubMedCentralCrossRef Browne SK, Zaman R, Sampaio EP, Jutivorakool K, Rosen LB, Ding L, Pancholi MJ, Yang LM, Priel DL, Uzel G, Freeman AF, Hayes CE et al (2012) Anti-CD20 (rituximab) therapy for anti–IFN-γ autoantibody–associated nontuberculous mycobacterial infection. Blood 119(17):3933–3939. doi:10.​1182/​blood-2011-12-395707 PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Rosen LB, Freeman AF, Yang LM, Jutivorakool K, Olivier KN, Angkasekwinai N, Suputtamongkol Y, Bennett JE, Pyrgos V, Williamson PR, Ding L, Holland SM et al (2013) Anti-granulocyte-macrophage colony stimulating factor autoantibodies in patients with cryptococcal meningitis. J Immunol 190(8):3959–3966. doi:10.4049/jimmunol.1202526 PubMedPubMedCentralCrossRef Rosen LB, Freeman AF, Yang LM, Jutivorakool K, Olivier KN, Angkasekwinai N, Suputtamongkol Y, Bennett JE, Pyrgos V, Williamson PR, Ding L, Holland SM et al (2013) Anti-granulocyte-macrophage colony stimulating factor autoantibodies in patients with cryptococcal meningitis. J Immunol 190(8):3959–3966. doi:10.​4049/​jimmunol.​1202526 PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Beerli RR, Bauer M, Fritzer A, Rosen LB, Buser RB, Hanner M, Maudrich M, Nebenfuehr M, Toepfer JAS, Mangold S, Bauer A, Holland SM et al (2014) Mining the human autoantibody repertoire: isolation of potent IL17A-neutralizing monoclonal antibodies from a patient with thymoma. MAbs 6(6):1608–1620. doi:10.4161/mabs.36292 PubMedPubMedCentralCrossRef Beerli RR, Bauer M, Fritzer A, Rosen LB, Buser RB, Hanner M, Maudrich M, Nebenfuehr M, Toepfer JAS, Mangold S, Bauer A, Holland SM et al (2014) Mining the human autoantibody repertoire: isolation of potent IL17A-neutralizing monoclonal antibodies from a patient with thymoma. MAbs 6(6):1608–1620. doi:10.​4161/​mabs.​36292 PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Branellec A, Bouillet L, Javaud N, Mekinian A, Boccon-Gibod I, Blanchard-Delaunay C, Oksenhendler E, Ollivier Y, Dunogué B, Amarger S, Ponard D, Drouet C et al (2012) Acquired C1-inhibitor deficiency: 7 patients treated with rituximab. J Clin Immunol 32(5):936–941. doi:10.1007/s10875-012-9691-2 PubMedCrossRef Branellec A, Bouillet L, Javaud N, Mekinian A, Boccon-Gibod I, Blanchard-Delaunay C, Oksenhendler E, Ollivier Y, Dunogué B, Amarger S, Ponard D, Drouet C et al (2012) Acquired C1-inhibitor deficiency: 7 patients treated with rituximab. J Clin Immunol 32(5):936–941. doi:10.​1007/​s10875-012-9691-2 PubMedCrossRef
76.
Zurück zum Zitat Lionet A, Provôt F, Glowacki F, Frémeaux-Bacchi V, Hazzan M (2009) A case of adult atypical haemolytic uraemic syndrome related to anti-factor H autoantibodies successfully treated by plasma exchange, corticosteroids and rituximab. NDT Plus 2(6):458–460. doi:10.1093/ndtplus/sfp109 PubMedPubMedCentral Lionet A, Provôt F, Glowacki F, Frémeaux-Bacchi V, Hazzan M (2009) A case of adult atypical haemolytic uraemic syndrome related to anti-factor H autoantibodies successfully treated by plasma exchange, corticosteroids and rituximab. NDT Plus 2(6):458–460. doi:10.​1093/​ndtplus/​sfp109 PubMedPubMedCentral
77.
Zurück zum Zitat Czaja CA, Merkel PA, Chan ED, Lenz LL, Wolf ML, Alam R, Frankel SK, Fischer A, Gogate S, Perez-Velez CM, Knight V (2014) Rituximab as successful adjunct treatment in a patient with disseminated nontuberculous mycobacterial infection due to acquired anti-interferon-γ autoantibody. Clin Infect Dis 58(6):e115–e118. doi:10.1093/cid/cit809 PubMedCrossRef Czaja CA, Merkel PA, Chan ED, Lenz LL, Wolf ML, Alam R, Frankel SK, Fischer A, Gogate S, Perez-Velez CM, Knight V (2014) Rituximab as successful adjunct treatment in a patient with disseminated nontuberculous mycobacterial infection due to acquired anti-interferon-γ autoantibody. Clin Infect Dis 58(6):e115–e118. doi:10.​1093/​cid/​cit809 PubMedCrossRef
78.
Zurück zum Zitat Sinha A, Gulati A, Saini S, Blanc C, Gupta A, Gurjar BS, Saini H, Kotresh ST, Ali U, Bhatia D, Ohri A, Kumar M et al (2014) Prompt plasma exchanges and immunosuppressive treatment improves the outcomes of anti-factor H autoantibody-associated hemolytic uremic syndrome in children. Kidney Int 85(5):1151–1160. doi:10.1038/ki.2013.373 PubMedCrossRef Sinha A, Gulati A, Saini S, Blanc C, Gupta A, Gurjar BS, Saini H, Kotresh ST, Ali U, Bhatia D, Ohri A, Kumar M et al (2014) Prompt plasma exchanges and immunosuppressive treatment improves the outcomes of anti-factor H autoantibody-associated hemolytic uremic syndrome in children. Kidney Int 85(5):1151–1160. doi:10.​1038/​ki.​2013.​373 PubMedCrossRef
80.
Zurück zum Zitat Marsh RA, Allen CE, McClain KL, Weinstein JL, Kanter J, Skiles J, Lee ND, Khan SP, Lawrence J, Mo JQ, Bleesing JJ, Filipovich AH, Jordan MB (2013) Salvage therapy of refractory hemophagocytic lymphohistiocytosis with alemtuzumab. Pediatr Blood Cancer 60(1):101–109. doi:10.1002/pbc.24188 PubMedCrossRef Marsh RA, Allen CE, McClain KL, Weinstein JL, Kanter J, Skiles J, Lee ND, Khan SP, Lawrence J, Mo JQ, Bleesing JJ, Filipovich AH, Jordan MB (2013) Salvage therapy of refractory hemophagocytic lymphohistiocytosis with alemtuzumab. Pediatr Blood Cancer 60(1):101–109. doi:10.​1002/​pbc.​24188 PubMedCrossRef
81.
Zurück zum Zitat Hillmen P, Young NS, Schubert J, Brodsky RA, Socié G, Muus P, Röth A, Szer J, Elebute MO, Nakamura R, Browne P, Risitano AM et al (2006) The complement inhibitor eculizumab in paroxysmal nocturnal hemoglobinuria. N Engl J Med 355(12):1233–1243. doi:10.1056/NEJMoa061648 PubMedCrossRef Hillmen P, Young NS, Schubert J, Brodsky RA, Socié G, Muus P, Röth A, Szer J, Elebute MO, Nakamura R, Browne P, Risitano AM et al (2006) The complement inhibitor eculizumab in paroxysmal nocturnal hemoglobinuria. N Engl J Med 355(12):1233–1243. doi:10.​1056/​NEJMoa061648 PubMedCrossRef
82.
Zurück zum Zitat Kelly RJ, Hill A, Arnold LM, Brooksbank GL, Richards SJ, Cullen M, Mitchell LD, Cohen DR, Gregory WM, Hillmen P (2011) Long-term treatment with eculizumab in paroxysmal nocturnal hemoglobinuria: sustained efficacy and improved survival. Blood 117(25):6786–6792. doi:10.1182/blood-2011-02-333997 PubMedCrossRef Kelly RJ, Hill A, Arnold LM, Brooksbank GL, Richards SJ, Cullen M, Mitchell LD, Cohen DR, Gregory WM, Hillmen P (2011) Long-term treatment with eculizumab in paroxysmal nocturnal hemoglobinuria: sustained efficacy and improved survival. Blood 117(25):6786–6792. doi:10.​1182/​blood-2011-02-333997 PubMedCrossRef
83.
Zurück zum Zitat Legendre CM, Licht C, Muus P, Greenbaum LA, Babu S, Bedrosian C, Bingham C, Cohen DJ, Delmas Y, Douglas K, Eitner F, Feldkamp T et al (2013) Terminal complement inhibitor eculizumab in atypical hemolytic-uremic syndrome. N Engl J Med 368(23):2169–2181. doi:10.1056/NEJMoa1208981 PubMedCrossRef Legendre CM, Licht C, Muus P, Greenbaum LA, Babu S, Bedrosian C, Bingham C, Cohen DJ, Delmas Y, Douglas K, Eitner F, Feldkamp T et al (2013) Terminal complement inhibitor eculizumab in atypical hemolytic-uremic syndrome. N Engl J Med 368(23):2169–2181. doi:10.​1056/​NEJMoa1208981 PubMedCrossRef
84.
Zurück zum Zitat Liu L, Okada S, Kong X-F, Kreins AY, Cypowyj S, Abhyankar A, Toubiana J, Itan Y, Audry M, Nitschke P, Masson C, Toth B et al (2011) Gain-of-function human STAT1 mutations impair IL-17 immunity and underlie chronic mucocutaneous candidiasis. J Exp Med 208(8):1635–1648. doi:10.1084/jem.20110958 PubMedPubMedCentralCrossRef Liu L, Okada S, Kong X-F, Kreins AY, Cypowyj S, Abhyankar A, Toubiana J, Itan Y, Audry M, Nitschke P, Masson C, Toth B et al (2011) Gain-of-function human STAT1 mutations impair IL-17 immunity and underlie chronic mucocutaneous candidiasis. J Exp Med 208(8):1635–1648. doi:10.​1084/​jem.​20110958 PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Higgins E, Al Shehri T, McAleer MA, Conlon N, Feighery C, Lilic D, Irvine AD (2015) Use of ruxolitinib to successfully treat chronic mucocutaneous candidiasis caused by gain-of-function signal transducer and activator of transcription 1 (STAT1) mutation. J Allergy Clin Immunol 135(2):551–553. doi:10.1016/j.jaci.2014.12.1867 PubMedCrossRef Higgins E, Al Shehri T, McAleer MA, Conlon N, Feighery C, Lilic D, Irvine AD (2015) Use of ruxolitinib to successfully treat chronic mucocutaneous candidiasis caused by gain-of-function signal transducer and activator of transcription 1 (STAT1) mutation. J Allergy Clin Immunol 135(2):551–553. doi:10.​1016/​j.​jaci.​2014.​12.​1867 PubMedCrossRef
86.
87.
Zurück zum Zitat Das R, Guan P, Sprague L, Verbist K, Tedrick P, An QA, Cheng C, Kurachi M, Levine R, Wherry EJ, Canna SW, Behrens EM, Nichols KE (2016) Janus kinase inhibition lessens inflammation and ameliorates disease in murine models of hemophagocytic lymphohistiocytosis. Blood 127(13):1666–1675. doi:10.1182/blood-2015-12-684399 PubMedCrossRef Das R, Guan P, Sprague L, Verbist K, Tedrick P, An QA, Cheng C, Kurachi M, Levine R, Wherry EJ, Canna SW, Behrens EM, Nichols KE (2016) Janus kinase inhibition lessens inflammation and ameliorates disease in murine models of hemophagocytic lymphohistiocytosis. Blood 127(13):1666–1675. doi:10.​1182/​blood-2015-12-684399 PubMedCrossRef
90.
91.
Zurück zum Zitat Bindl L, Torgerson T, Perroni L, Youssef N, Ochs HD, Goulet O, Ruemmele FM (2005) Successful use of the new immune-suppressor sirolimus in IPEX (immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome). J Pediatr 147(2):256–259. doi:10.1016/j.jpeds.2005.04.017 PubMedCrossRef Bindl L, Torgerson T, Perroni L, Youssef N, Ochs HD, Goulet O, Ruemmele FM (2005) Successful use of the new immune-suppressor sirolimus in IPEX (immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome). J Pediatr 147(2):256–259. doi:10.​1016/​j.​jpeds.​2005.​04.​017 PubMedCrossRef
93.
94.
Zurück zum Zitat Angulo I, Vadas O, Garçon F, Banham-Hall E, Plagnol V, Leahy TR, Baxendale H, Coulter T, Curtis J, Wu C, Blake-Palmer K, Perisic O et al (2013) Phosphoinositide 3-kinase δ gene mutation predisposes to respiratory infection and airway damage. Science 342(6160):866–871. doi:10.1126/science.1243292 PubMedPubMedCentralCrossRef Angulo I, Vadas O, Garçon F, Banham-Hall E, Plagnol V, Leahy TR, Baxendale H, Coulter T, Curtis J, Wu C, Blake-Palmer K, Perisic O et al (2013) Phosphoinositide 3-kinase δ gene mutation predisposes to respiratory infection and airway damage. Science 342(6160):866–871. doi:10.​1126/​science.​1243292 PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Lucas CL, Kuehn HS, Zhao F, Niemela JE, Deenick EK, Palendira U, Avery DT, Moens L, Cannons JL, Biancalana M, Stoddard J, Ouyang W et al (2014) Dominant-activating germline mutations in the gene encoding the PI(3)K catalytic subunit p110δ result in T cell senescence and human immunodeficiency. Nat Immunol 15(1):88–97. doi:10.1038/ni.2771 PubMedCrossRef Lucas CL, Kuehn HS, Zhao F, Niemela JE, Deenick EK, Palendira U, Avery DT, Moens L, Cannons JL, Biancalana M, Stoddard J, Ouyang W et al (2014) Dominant-activating germline mutations in the gene encoding the PI(3)K catalytic subunit p110δ result in T cell senescence and human immunodeficiency. Nat Immunol 15(1):88–97. doi:10.​1038/​ni.​2771 PubMedCrossRef
96.
Zurück zum Zitat Harris J, Hartman M, Roche C, Zeng SG, O’Shea A, Sharp FA, Lambe EM, Creagh EM, Golenbock DT, Tschopp J, Kornfeld H, Fitzgerald KA et al (2011) Autophagy controls IL-1β secretion by targeting pro-IL-1β for degradation. J Biol Chem 286(11):9587–9597. doi:10.1074/jbc.M110.202911 PubMedPubMedCentralCrossRef Harris J, Hartman M, Roche C, Zeng SG, O’Shea A, Sharp FA, Lambe EM, Creagh EM, Golenbock DT, Tschopp J, Kornfeld H, Fitzgerald KA et al (2011) Autophagy controls IL-1β secretion by targeting pro-IL-1β for degradation. J Biol Chem 286(11):9587–9597. doi:10.​1074/​jbc.​M110.​202911 PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Bigorgne AE, Farin HF, Lemoine R, Mahlaoui N, Lambert N, Gil M, Schulz A, Philippet P, Schlesser P, Abrahamsen TG, Oymar K, Davies EG et al (2014) TTC7A mutations disrupt intestinal epithelial apicobasal polarity. J Clin Invest 124(1):328–337. doi:10.1172/JCI71471 PubMedCrossRef Bigorgne AE, Farin HF, Lemoine R, Mahlaoui N, Lambert N, Gil M, Schulz A, Philippet P, Schlesser P, Abrahamsen TG, Oymar K, Davies EG et al (2014) TTC7A mutations disrupt intestinal epithelial apicobasal polarity. J Clin Invest 124(1):328–337. doi:10.​1172/​JCI71471 PubMedCrossRef
99.
Zurück zum Zitat Chan B, Wara D, Bastian J, Hershfield MS, Bohnsack J, Azen CG, Parkman R, Weinberg K, Kohn DB (2005) Long-term efficacy of enzyme replacement therapy for adenosine deaminase (ADA)-deficient severe combined immunodeficiency (SCID). Clin Immunol 117(2):133–143. doi:10.1016/j.clim.2005.07.006 PubMedCrossRef Chan B, Wara D, Bastian J, Hershfield MS, Bohnsack J, Azen CG, Parkman R, Weinberg K, Kohn DB (2005) Long-term efficacy of enzyme replacement therapy for adenosine deaminase (ADA)-deficient severe combined immunodeficiency (SCID). Clin Immunol 117(2):133–143. doi:10.​1016/​j.​clim.​2005.​07.​006 PubMedCrossRef
100.
Zurück zum Zitat Serana F, Sottini A, Chiarini M, Zanotti C, Ghidini C, Lanfranchi A, Notarangelo LD, Caimi L, Imberti L (2010) The different extent of B and T cell immune reconstitution after hematopoietic stem cell transplantation and enzyme replacement therapies in SCID patients with adenosine deaminase deficiency. J Immunol 185(12):7713–7722. doi:10.4049/jimmunol.1001770 PubMedCrossRef Serana F, Sottini A, Chiarini M, Zanotti C, Ghidini C, Lanfranchi A, Notarangelo LD, Caimi L, Imberti L (2010) The different extent of B and T cell immune reconstitution after hematopoietic stem cell transplantation and enzyme replacement therapies in SCID patients with adenosine deaminase deficiency. J Immunol 185(12):7713–7722. doi:10.​4049/​jimmunol.​1001770 PubMedCrossRef
102.
Zurück zum Zitat Krassilnikova S, Craig ET, Craig TJ (2010) Summary of the International Multicenter Prospective Angioedema C1-inhibitor Trials 1 and 2 (IMPACT1 and 2). Expert Rev Clin Immunol 6(3):327–334. doi:10.1586/eci.10.19 PubMedCrossRef Krassilnikova S, Craig ET, Craig TJ (2010) Summary of the International Multicenter Prospective Angioedema C1-inhibitor Trials 1 and 2 (IMPACT1 and 2). Expert Rev Clin Immunol 6(3):327–334. doi:10.​1586/​eci.​10.​19 PubMedCrossRef
103.
Zurück zum Zitat Lumry W, Manning ME, Hurewitz DS, Davis-Lorton M, Fitts D, Kalfus IN, Uknis ME (2013) Nanofiltered C1-esterase inhibitor for the acute management and prevention of hereditary angioedema attacks due to C1-inhibitor deficiency in children. J Pediatr 162(5):1017–1022. doi:10.1016/j.jpeds.2012.11.030 PubMedCrossRef Lumry W, Manning ME, Hurewitz DS, Davis-Lorton M, Fitts D, Kalfus IN, Uknis ME (2013) Nanofiltered C1-esterase inhibitor for the acute management and prevention of hereditary angioedema attacks due to C1-inhibitor deficiency in children. J Pediatr 162(5):1017–1022. doi:10.​1016/​j.​jpeds.​2012.​11.​030 PubMedCrossRef
104.
Zurück zum Zitat Manson AL, Dempster J, Grigoriadou S, Buckland MS, Longhurst HJ (2014) Use of recombinant C1 inhibitor in patients with resistant or frequent attacks of hereditary or acquired angioedema. Eur J Dermatol 24(1):28–34. doi:10.1684/ejd.2013.2252 PubMed Manson AL, Dempster J, Grigoriadou S, Buckland MS, Longhurst HJ (2014) Use of recombinant C1 inhibitor in patients with resistant or frequent attacks of hereditary or acquired angioedema. Eur J Dermatol 24(1):28–34. doi:10.​1684/​ejd.​2013.​2252 PubMed
106.
Zurück zum Zitat Chaigne-Delalande B, Li F-Y, O’Connor GM, Lukacs MJ, Jiang P, Zheng L, Shatzer A, Biancalana M, Pittaluga S, Matthews HF, Jancel TJ, Bleesing JJ et al (2013) Mg2+ regulates cytotoxic functions of NK and CD8 T cells in chronic EBV infection through NKG2D. Science 341(6142):186–191. doi:10.1126/science.1240094 PubMedPubMedCentralCrossRef Chaigne-Delalande B, Li F-Y, O’Connor GM, Lukacs MJ, Jiang P, Zheng L, Shatzer A, Biancalana M, Pittaluga S, Matthews HF, Jancel TJ, Bleesing JJ et al (2013) Mg2+ regulates cytotoxic functions of NK and CD8 T cells in chronic EBV infection through NKG2D. Science 341(6142):186–191. doi:10.​1126/​science.​1240094 PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Fernandez-Boyanapalli RF, Frasch SC, Thomas SM, Malcolm KC, Nicks M, Harbeck RJ, Jakubzick CV, Nemenoff R, Henson PM, Holland SM, Bratton DL (2015) Pioglitazone restores phagocyte mitochondrial oxidants and bactericidal capacity in chronic granulomatous disease. J Allergy Clin Immunol 135(2):517–527 . doi:10.1016/j.jaci.2014.10.034e12PubMedCrossRef Fernandez-Boyanapalli RF, Frasch SC, Thomas SM, Malcolm KC, Nicks M, Harbeck RJ, Jakubzick CV, Nemenoff R, Henson PM, Holland SM, Bratton DL (2015) Pioglitazone restores phagocyte mitochondrial oxidants and bactericidal capacity in chronic granulomatous disease. J Allergy Clin Immunol 135(2):517–527 . doi:10.​1016/​j.​jaci.​2014.​10.​034e12PubMedCrossRef
109.
Zurück zum Zitat Migliavacca M, Assanelli A, Ferrua F, Cicalese MP, Biffi A, Frittoli M, Silvani P, Chidini G, Calderini E, Mandelli A, Camporesi A, Milani R et al (2016) Pioglitazone as a novel therapeutic approach in chronic granulomatous disease. J Allergy Clin Immunol. doi:10.1016/j.jaci.2016.01.033 PubMedPubMedCentral Migliavacca M, Assanelli A, Ferrua F, Cicalese MP, Biffi A, Frittoli M, Silvani P, Chidini G, Calderini E, Mandelli A, Camporesi A, Milani R et al (2016) Pioglitazone as a novel therapeutic approach in chronic granulomatous disease. J Allergy Clin Immunol. doi:10.​1016/​j.​jaci.​2016.​01.​033 PubMedPubMedCentral
110.
Zurück zum Zitat Dowdell KC, Pesnicak L, Hoffmann V, Steadman K, Remaley AT, Cohen JI, Straus SE, Rao VK (2009) Valproic acid (VPA), a histone deacetylase (HDAC) inhibitor, diminishes lymphoproliferation in the Fas-deficient MRL/lpr(−/−) murine model of autoimmune lymphoproliferative syndrome (ALPS). Exp Hematol 37(4):487–494. doi:10.1016/j.exphem.2008.12.002 PubMedPubMedCentralCrossRef Dowdell KC, Pesnicak L, Hoffmann V, Steadman K, Remaley AT, Cohen JI, Straus SE, Rao VK (2009) Valproic acid (VPA), a histone deacetylase (HDAC) inhibitor, diminishes lymphoproliferation in the Fas-deficient MRL/lpr(−/−) murine model of autoimmune lymphoproliferative syndrome (ALPS). Exp Hematol 37(4):487–494. doi:10.​1016/​j.​exphem.​2008.​12.​002 PubMedPubMedCentralCrossRef
111.
Zurück zum Zitat Bobé P, Bonardelle D, Benihoud K, Opolon P, Chelbi-Alix MK (2006) Arsenic trioxide: a promising novel therapeutic agent for lymphoproliferative and autoimmune syndromes in MRL/lpr mice. Blood 108(13):3967–3975. doi:10.1182/blood-2006-04-020610 PubMedCrossRef Bobé P, Bonardelle D, Benihoud K, Opolon P, Chelbi-Alix MK (2006) Arsenic trioxide: a promising novel therapeutic agent for lymphoproliferative and autoimmune syndromes in MRL/lpr mice. Blood 108(13):3967–3975. doi:10.​1182/​blood-2006-04-020610 PubMedCrossRef
114.
Zurück zum Zitat Coll RC, Robertson AAB, Chae JJ, Higgins SC, Muñoz-Planillo R, Inserra MC, Vetter I, Dungan LS, Monks BG, Stutz A, Croker DE, Butler MS et al (2015) A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat Med 21(3):248–255. doi:10.1038/nm.3806 PubMedPubMedCentral Coll RC, Robertson AAB, Chae JJ, Higgins SC, Muñoz-Planillo R, Inserra MC, Vetter I, Dungan LS, Monks BG, Stutz A, Croker DE, Butler MS et al (2015) A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat Med 21(3):248–255. doi:10.​1038/​nm.​3806 PubMedPubMedCentral
115.
Zurück zum Zitat Youm YH, Nguyen KY, Grant RW, Goldberg EL, Bodogai M, Kim D, D’Agostino D, Planavsky N, Lupfer C, Kanneganti TD, Kang S, Horvath TL et al (2015) The ketone metabolite β-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med 21(3):263–269. doi:10.1038/nm.3804 PubMedPubMedCentral Youm YH, Nguyen KY, Grant RW, Goldberg EL, Bodogai M, Kim D, D’Agostino D, Planavsky N, Lupfer C, Kanneganti TD, Kang S, Horvath TL et al (2015) The ketone metabolite β-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med 21(3):263–269. doi:10.​1038/​nm.​3804 PubMedPubMedCentral
117.
Zurück zum Zitat Joseph K, Tholanikunnel BG, Bygum A, Ghebrehiwet B, Kaplan AP (2013) Factor XII-independent activation of the bradykinin-forming cascade: implications for the pathogenesis of hereditary angioedema types I and II. J Allergy Clin Immunol 132(2):470–475. doi:10.1016/j.jaci.2013.03.026 PubMedCrossRef Joseph K, Tholanikunnel BG, Bygum A, Ghebrehiwet B, Kaplan AP (2013) Factor XII-independent activation of the bradykinin-forming cascade: implications for the pathogenesis of hereditary angioedema types I and II. J Allergy Clin Immunol 132(2):470–475. doi:10.​1016/​j.​jaci.​2013.​03.​026 PubMedCrossRef
118.
Zurück zum Zitat Wintenberger C, Boccon-Gibod I, Launay D, Fain O, Kanny G, Jeandel PY, Martin L, Gompel A, Bouillet L (2014) Tranexamic acid as maintenance treatment for non-histaminergic angioedema: analysis of efficacy and safety in 37 patients. Clin Exp Immunol 178(1):112–117. doi:10.1111/cei.12379 PubMedPubMedCentralCrossRef Wintenberger C, Boccon-Gibod I, Launay D, Fain O, Kanny G, Jeandel PY, Martin L, Gompel A, Bouillet L (2014) Tranexamic acid as maintenance treatment for non-histaminergic angioedema: analysis of efficacy and safety in 37 patients. Clin Exp Immunol 178(1):112–117. doi:10.​1111/​cei.​12379 PubMedPubMedCentralCrossRef
120.
Zurück zum Zitat Farkas H, Harmat G, Füst G, Varga L, Visy B (2002) Clinical management of hereditary angio-oedema in children. Pediatr Allergy Immunol 13(3):153–161PubMedCrossRef Farkas H, Harmat G, Füst G, Varga L, Visy B (2002) Clinical management of hereditary angio-oedema in children. Pediatr Allergy Immunol 13(3):153–161PubMedCrossRef
121.
Zurück zum Zitat Vece TJ, Watkin LB, Nicholas SK, Canter D, Braun MC, Guillerman RP, Eldin KW, Bertolet G, McKinley SD, de Guzman M, Forbes LR, Chinn I et al (2016) Copa syndrome: a novel autosomal dominant immune dysregulatory disease. J Clin Immunol 36(4):377–387. doi:10.1007/s10875-016-0271-8 PubMedCrossRef Vece TJ, Watkin LB, Nicholas SK, Canter D, Braun MC, Guillerman RP, Eldin KW, Bertolet G, McKinley SD, de Guzman M, Forbes LR, Chinn I et al (2016) Copa syndrome: a novel autosomal dominant immune dysregulatory disease. J Clin Immunol 36(4):377–387. doi:10.​1007/​s10875-016-0271-8 PubMedCrossRef
Metadaten
Titel
An Update on the Use of Immunomodulators in Primary Immunodeficiencies
Publikationsdatum
21.11.2016
Erschienen in
Clinical Reviews in Allergy & Immunology / Ausgabe 2/2017
Print ISSN: 1080-0549
Elektronische ISSN: 1559-0267
DOI
https://doi.org/10.1007/s12016-016-8591-2

Weitere Artikel der Ausgabe 2/2017

Clinical Reviews in Allergy & Immunology 2/2017 Zur Ausgabe

Update HNO

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