218
Views
5
CrossRef citations to date
0
Altmetric
Review

Safety of systemic treatments for Behçet’s syndrome

ORCID Icon, ORCID Icon, , , , , , , ORCID Icon & show all
Pages 1269-1301 | Received 23 Jun 2020, Accepted 25 Aug 2020, Published online: 15 Sep 2020

References

  • Lopalco G, Cantarini L, Vitale A, et al. Interleukin-1 as a common denominator from autoinflammatory to autoimmune disorders: premises, perils, and perspectives. Mediators Inflamm. 2015;2015:194864.
  • Gül A. Behçet’s disease as an autoinflammatory disorder. Curr Drug Targets Inflamm Allergy. 2005;4(1):81–83.
  • Jennette JC, Falk RJ, Bacon PA, et al. 2012 revised international chapel hill consensus conference nomenclature of vasculitides. Arthritis Rheum. 2013;65(1):1–11.
  • Lopalco G, Lucherini OM, Vitale A, et al. Putative role of serum amyloid-A and proinflammatory cytokines as biomarkers for Behçet’s disease. Medicine (Baltimore). 2015;94(42):e1858.
  • Hamzaoui K, Hamzaoui A, Guemira F, et al. Cytokine profile in Behçet’s disease patients. Relationship with disease activity. Scand J Rheumatol. 2002;31(4):205–210.
  • Lopalco G, Lucherini OM, Lopalco A, et al. Cytokine signatures in mucocutaneous and ocular Behçet’s disease. Front Immunol. 2017;8:200.
  • Rigante D, Vitale A, Natale MF, et al. A comprehensive comparison between pediatric and adult patients with periodic fever, aphthous stomatitis, pharyngitis, and cervical adenopathy (PFAPA) syndrome. Clin Rheumatol. 2017;36:463–468.
  • Cattalini M, Soliani M, Rigante D, et al. Basic characteristics of adults with periodic fever, aphthous stomatitis, pharyngitis, and adenopathy syndrome in comparison with the typical pediatric expression of disease. Mediators Inflamm. 2015;2015:570418.
  • Cantarini L, Vitale A, Bartolomei B, et al. Diagnosis of PFAPA syndrome applied to a cohort of 17 adults with unexplained recurrent fevers. Clin Exp Rheumatol. 2012;30(2):269–271.
  • Arida A, Sfikakis PP. Anti-cytokine biologic treatment beyond anti-TNF in Behçet’s disease. Clin Exp Rheumatol. 2014;32(4 Suppl 84):S149‐S155.
  • Rotondo C, Lopalco G, Iannone F, et al. Mucocutaneous involvement in Behçet’s disease: how systemic treatment has changed in the last decades and future perspectives. Mediators Inflamm. 2015;2015:451675.
  • Emmi G, Silvestri E, Squatrito D, et al. Tocilizumab-induced exacerbation of mucosal ulcers in a patient with multi-refractory Behçet׳s disease. Semin Arthritis Rheum. 2016;46(1):e1–e2.
  • Yazici H, Seyahi E, Hatemi G, et al. Behçet syndrome: a contemporary view. Nat Rev Rheumatol. 2018;14(2):107–119. [published correction appears in Nat Rev Rheumatol. 2018 Jan 24;14 (2): 119].
  • Alpsoy E, Zouboulis CC, Ehrlich GE. Mucocutaneous lesions of Behçet’s disease. Yonsei Med J. 2007;48(4):573–585.
  • Hatemi G, Christensen R, Bang D, et al. update of the EULAR recommendations for the management of Behçet’s syndrome. Ann Rheum Dis. 2018;77(6):808–818.
  • Yazici Y, Yurdakul S, Yazici H. Behçet’s syndrome. Curr Rheumatol Rep. 2010;12(6):429–435.
  • Yazici H, Fresko I, Yurdakul S. Behçet’s syndrome: disease manifestations, management, and advances in treatment. Nat Clin Pract Rheumatol. 2007;3(3):148–155.
  • Choi IJ, Kim JS, Cha SD, et al. Long-term clinical course and prognostic factors in intestinal Behçet’s disease. Dis Colon Rectum. 2000;43(5):692–700.
  • Moral F, Hamuryudan V, Yurdakul S, et al. Inefficacy of azapropazone in the acute arthritis of Behçet’s syndrome: a randomized, double blind, placebo controlled study. Clin Exp Rheumatol. 1995;13(4):493–495.
  • Szekanecz Z, Szántó S, Szabó Z, et al. Biologics - beyond the joints. Autoimmun Rev. 2010;9(12):820–824.
  • Tüzün H, Beşirli K, Sayin A, et al. Management of aneurysms in Behçet’s syndrome: an analysis of 24 patients. Surgery. 1997;121(2):150–156.
  • Akman-Demir G, Serdaroglu P, Tasçi B. Clinical patterns of neurological involvement in Behçet’s disease: evaluation of 200 patients. The Neuro-Behçet Study Group. Brain. 1999;122(Pt 11):2171–2182.
  • Wee JS, Marinaki A, Smith CH. Life threatening myelotoxicity secondary to azathioprine in a patient with atopic eczema and normal thiopurine methyltransferase activity. BMJ. 2011;342:d1417.
  • Relling MV, Gardner EE, Sandborn WJ, et al. Clinical pharmacogenetics implementation consortium guidelines for thiopurine methyltransferase genotype and thiopurine dosing: 2013 update. Clin Pharmacol Ther. 2013;93(4):324–325.
  • Gearry RB, Day AS, Barclay ML, et al. Azathioprine and allopurinol: A two-edged interaction. J Gastroenterol Hepatol. 2010;25(4):653–655.
  • Vazquez SR, Rondina MT, Pendleton RC. Azathioprine-induced warfarin resistance. Ann Pharmacother. 2008;42(7):1118–1123.
  • Yazici H, Pazarli H, Barnes CG, et al. A controlled trial of azathioprine in Behçet’s syndrome. N Engl J Med. 1990;322(5):281–285.
  • Hamuryudan V, Ozyazgan Y, Hizli N, et al. Azathioprine in Behcet’s syndrome: effects on long-term prognosis. Arthritis Rheum. 1997;40(4):769–774.
  • Evereklioglu C. Current concepts in the etiology and treatment of Behçet disease. Surv Ophthalmol. 2005;50(4):297–350.
  • Hatemi I, Esatoglu SN, Hatemi G, et al. Characteristics, treatment, and long-term outcome of gastrointestinal involvement in Behçet’s syndrome: a strobe-compliant observational study from a dedicated multidisciplinary center. Medicine (Baltimore). 2016;95(16):e3348.
  • Davatchi F, Sadeghi Abdollahi B, Shams H, et al. Combination of pulse cyclophosphamide and azathioprine in ocular manifestations of Behçet’s disease: longitudinal study of up to 10 years. Int J Rheum Dis. 2014;17(4):444–452.
  • Petri M. Immunosuppressive drug use in pregnancy. Autoimmunity. 2003;36(1):51–56.
  • Nguyen GC, Seow CH, Maxwell C, et al. The Toronto consensus statements for the management of inflammatory bowel disease in pregnancy. Gastroenterology. 2016;150(3):734–757.e1.
  • Christensen LA, Dahlerup JF, Nielsen MJ, et al. Azathioprine treatment during lactation. Aliment Pharmacol Ther. 2008;28(10):1209–1213.
  • Bar-Gil Shitrit A, Grisaru-Granovsky S, Ben Ya’acov A, et al. Management of inflammatory bowel disease during pregnancy. Dig Dis Sci. 2016;61(8):2194–2204.
  • Matsumura N, Mizushima Y. Leucocyte movement and colchicine treatment in Behcet’s disease. Lancet. 1975;2(7939):813.
  • Aktulga E, Altaç M, Müftüoglu A, et al. A double-blind study of colchicine in Behçet’s disease. Haematologica. 1980;65(3):399–402.
  • Yurdakul S, Mat C, Tüzün Y, et al. A double-blind trial of colchicine in Behçet’s syndrome. Arthritis Rheum. 2001;44(11):2686–2692.
  • Masuda K, Nakajima A, Urayama A, et al. Double-masked trial of cyclosporin versus colchicine and long-term open study of cyclosporin in Behçet’s disease. Lancet. 1989;1(8647):1093–1096.
  • Gürler A, Boyvat A, Türsen U. Clinical manifestations of Behçet’s disease: an analysis of 2147 patients. Yonsei Med J. 1997;38(6):423–427.
  • Tursen U, Gurler A, Boyvat A. Evaluation of clinical findings according to sex in 2313 Turkish patients with Behçet’s disease. Int J Dermatol. 2003;42(5):346–351.
  • Davatchi F, Sadeghi Abdollahi B, Tehrani Banihashemi A, et al. Colchicine versus placebo in Behçet’s disease: randomized double-blind controlled cross-over trial. Mod Rheumatol. 2009;19(5):542–549.
  • Finkelstein Y, Aks SE, Hutson JR, et al. Colchicine poisoning: the dark side of an ancient drug. Clin Toxicol (Phila). 2010;48(5):407–414.
  • Aghabiklooei A, Zamani N, Hassanian-Moghaddam H, et al. Acute colchicine overdose: report of three cases. Reumatismo. 2014;65(6):307–311.
  • Kuncl RW, Duncan G, Watson D, et al. Colchicine myopathy and neuropathy. N Engl J Med. 1987;316(25):1562–1568.
  • Boonmuang P, Nathisuwan S, Chaiyakunapruk N, et al. Characterization of statin-associated myopathy case reports in thailand using the health product vigilance center database. Drug Saf. 2013;36(9):779–787. [published correction appears in Drug Saf. 2013 Jul;36(7):583-4].
  • Michael O, Goldman RD, Koren G. Motherisk Team. Safety of colchicine therapy during pregnancy. Can Fam Physician. 2003;49:967–969.
  • Mizushima Y, Matsumura N, Mori M, et al. Colchicine in Behçet’s disease. Lancet. 1977;2(8046):1037.
  • Haimov-Kochman R, Ben-Chetrit E. The effect of colchicine treatment on sperm production and function: a review. Hum Reprod. 1998;13(2):360–362.
  • Sammaritano LR, Bermas BL, Chakravarty EE, et al. American college of rheumatology guideline for the management of reproductive health in rheumatic and musculoskeletal diseases. Arthritis Rheumatol. 2020;72(4):529–556.
  • Ramamoorthy S, Cidlowski JA. Corticosteroids: mechanisms of action in health and disease. Rheum Dis Clin North Am. 2016;42(1):15–vii.
  • Mat C, Yurdakul S, Uysal S, et al. A double-blind trial of depot corticosteroids in Behçet’s syndrome. Rheumatology (Oxford). 2006;45(3):348–352.
  • Hengge UR, Ruzicka T, Schwartz RA, et al. Adverse effects of topical glucocorticosteroids. J Am Acad Dermatol. 2006;54(1):1–18.
  • Mohammadi M, Shahram F, Shams H, et al. High-dose intravenous steroid pulse therapy in ocular involvement of Behçet’s disease: a pilot double-blind controlled study. Int J Rheum Dis. 2017;20(9):1269–1276.
  • Hamuryudan V, Yurdakul S, Moral F, et al. Pulmonary arterial aneurysms in Behçet’s syndrome: a report of 24 cases. Br J Rheumatol. 1994;33(1):48–51.
  • Park-Wyllie L, Mazzotta P, Pastuszak A, et al. Birth defects after maternal exposure to corticosteroids: prospective cohort study and meta-analysis of epidemiological studies. Teratology. 2000;62(6):385–392.
  • Ost L, Wettrell G, Björkhem I, et al. Prednisolone excretion in human milk. J Pediatr. 1985;106(6):1008–1011.
  • Hartvig P, Simonsson B, Oberg G, et al. Inter- and intraindividual differences in oral chlorambucil pharmacokinetics. Eur J Clin Pharmacol. 1988;35(5):551–554.
  • Abdalla MI, el-D Bahoat N. Long-lasting remission of Behçet’s disease after chlorambucil therapy. Br J Ophthalmol. 1973;57(9):706–711.
  • Mudun BA, Ergen A, Ipcioglu SU, et al. Short-term chlorambucil for refractory uveitis in Behçet’s disease. Ocul Immunol Inflamm. 2001;9(4):219–229.
  • Goldstein DA, Fontanilla FA, Kaul S, et al. Long-term follow-up of patients treated with short-term high-dose chlorambucil for sight-threatening ocular inflammation. Ophthalmology. 2002;109(2):370–377.
  • Elliott JH, Ballinger WH. Behçet’s syndrome: treatment with chlorambucil. Trans Am Ophthalmol Soc. 1984;82:264–281.
  • Tabbara KF. Chlorambucil in Behçet’s disease. A reappraisal. Ophthalmology. 1983;90(8):906–908.
  • Palmer RG, Denman AM. Malignancies induced by chlorambucil. Cancer Treat Rev. 1984;11(2):121–129.
  • Miserocchi E, Baltatzis S, Ekong A, et al. Efficacy and safety of chlorambucil in intractable noninfectious uveitis: the massachusetts eye and ear infirmary experience. Ophthalmology. 2002;109(1):137–142.
  • Zaghetto JM, Yamamoto MM, Souza MB, et al. Chlorambucil and cyclosporine A in Brazilian patients with Behçet’s disease uveitis: a retrospective study. Arq Bras Oftalmol. 2010;73(1):40–46.
  • Steege JF, Caldwell DS. Renal agenesis after first trimester exposure to chlorambucil. South Med J. 1980;73(10):1414–1415.
  • Mills KA, Chess-Williams R, McDermott C. Novel insights into the mechanism of cyclophosphamide-induced bladder toxicity: chloroacetaldehyde’s contribution to urothelial dysfunction in vitro. Arch Toxicol. 2019;93(11):3291–3303.
  • Ahlmann M, Hempel G. The effect of cyclophosphamide on the immune system: implications for clinical cancer therapy. Cancer Chemother Pharmacol. 2016;78(4):661–671.
  • Cavallasca JA, Costa CA, Maliandi Mdel R, et al. Severe infections in patients with autoimmune diseases treated with cyclophosphamide. Reumatol Clin. 2015;11(4):221–223.
  • Lopalco G, Rigante D, Venerito V, et al. Management of small vessel vasculitides. Curr Rheumatol Rep. 2016;18(6):36.
  • Fukutani K, Ishida H, Shinohara M, et al. Suppression of spermatogenesis in patients with Behçet’s disease treated with cyclophosphamide and colchicine. Fertil Steril. 1981;36(1):76–80.
  • Dan D, Fischer R, Adler S, et al. Cyclophosphamide: as bad as its reputation? Long-term single centre experience of cyclophosphamide side effects in the treatment of systemic autoimmune diseases. Swiss Med Wkly. 2014;144:w14030.
  • Ozyazgan Y, Yurdakul S, Yazici H, et al. Low dose cyclosporin A versus pulsed cyclophosphamide in Behçet’s syndrome: a single masked trial. Br J Ophthalmol. 1992;76(4):241–243.
  • Gurcan M, Esatoglu SN, Hamuryudan V, et al. Long term follow-up of Behçet’s syndrome patients treated with cyclophosphamide. Rheumatology (Oxford). 2019;kez598. [published online ahead of print, 2019 Dec 16]. doi:https://doi.org/10.1093/rheumatology/kez598
  • Martin F, Lauwerys B, Lefèbvre C, et al. Side-effects of intravenous cyclophosphamide pulse therapy. Lupus. 1997;6(3):254–257.
  • Monach PA, Arnold LM, Merkel PA. Incidence and prevention of bladder toxicity from cyclophosphamide in the treatment of rheumatic diseases: a data-driven review. Arthritis Rheum. 2010;62(1):9–21.
  • Noel N, Bernard R, Wechsler B, et al. Long-term outcome of neuro-Behçet’s disease. Arthritis Rheumatol. 2014;66(5):1306–1314.
  • Campagne O, Zhong B, Nair S, et al. Exposure-toxicity association of cyclophosphamide and its metabolites in infants and young children with primary brain tumors: implications for dosing. Clin Cancer Res. 2020;26(7):1563–1573.
  • Liddicoat AM, Lavelle EC. Modulation of innate immunity by cyclosporine A. Biochem Pharmacol. 2019;163:472–480.
  • Rodicio JL. Calcium antagonists and renal protection from cyclosporine nephrotoxicity: long-term trial in renal transplantation patients. J Cardiovasc Pharmacol. 2000;35(3 Suppl 1):S7–S11.
  • Díaz-Llopis M, Cervera M, Menezo JL. Cyclosporin A treatment of Behçet’s disease: a long-term study. Curr Eye Res. 1990;9 Suppl:17–23.
  • Takahashi T, Kamimura A. Cyclosporin A promotes hair epithelial cell proliferation and modulates protein kinase C expression and translocation in hair epithelial cells. J Invest Dermatol. 2001;117(3):605–611.
  • Pisanty S, Shoshan S, Chajek T, et al. The effect of cyclosporin A (CsA) treatment on gingival tissue of patients with Behçet’s disease. J Periodontol. 1988;59(9):599–603.
  • Ponnaiyan D, Jegadeesan V. Cyclosporine A: novel concepts in its role in drug-induced gingival overgrowth. Dent Res J (Isfahan). 2015;12(6):499–506.
  • Remuzzi G, Bertani T. Renal vascular and thrombotic effects of cyclosporine. Am J Kidney Dis. 1989;13:261–272.
  • Hinchey J, Chaves C, Appignani B, et al. A reversible posterior leukoencephalopathy syndrome. N Engl J Med. 1996;334(8):494–500.
  • Kotake S, Higashi K, Yoshikawa K, et al. Central nervous system symptoms in patients with Behçet disease receiving cyclosporine therapy. Ophthalmology. 1999;106(3):586–589.
  • Kötter I, Günaydin I, Batra M, et al. CNS involvement occurs more frequently in patients with Behçet’s disease under cyclosporin A (CSA) than under other medications–results of a retrospective analysis of 117 cases. Clin Rheumatol. 2006;25(4):482–486.
  • Akman-Demir G, Ayranci O, Kurtuncu M, et al. Cyclosporine for Behçet’s uveitis: is it associated with an increased risk of neurological involvement? Clin Exp Rheumatol. 2008;26(4 Suppl 50):S84–S90.
  • Reggia R, Bazzani C, Andreoli L, et al. The efficacy and safety of cyclosporin A in pregnant patients with systemic autoimmune diseases. Am J Reprod Immunol. 2016;75(6):654–660.
  • Galijasevic S. The development of myeloperoxidase inhibitors. Bioorg Med Chem Lett. 2019;29(1):1–7.
  • Sharquie KE, Najim RA, Abu-Raghif AR. Dapsone in Behçet’s disease: a double-blind placebo-controlled cross-over study. J Dermatol. 2002;29(5):267–279.
  • Sharquie KE. Suppression of Behçet’s disease with dapsone. Br J Dermatol. 1984;110(4):493–494.
  • Hamza M, Hamzaoui K, Ayed K. Treatment of Behçet’s disease with dapsone. Clin Rheumatol. 1989;8(1):113–114.
  • Lorenz M, Wozel G, Schmitt J. Hypersensitivity reactions to dapsone: a systematic review. Acta Derm Venereol. 2012;92(2):194–199.
  • Webster GF. Is topical dapsone safe in glucose-6-phosphate dehydrogenase-deficient and sulfonamide-allergic patients? J Drugs Dermatol. 2010;9(5):532–536.
  • Sanders SW, Zone JJ, Foltz RL, et al. Hemolytic anemia induced by dapsone transmitted through breast milk. Ann Intern Med. 1982;96(4):465–466.
  • Braun J, Rau R. An update on methotrexate. Curr Opin Rheumatol. 2009;21(3):216–223.
  • Haskó G, Cronstein BN. Adenosine: an endogenous regulator of innate immunity. Trends Immunol. 2004;25(1):33–39.
  • Iwata S, Saito K, Yamaoka K, et al. Efficacy of combination therapy of anti-TNF-α antibody infliximab and methotrexate in refractory entero-Behçet’s disease. Mod Rheumatol. 2011;21(2):184–191.
  • Bae JH, Lee SC. Effect of intravitreal methotrexate and aqueous humor cytokine levels in refractory retinal vasculitis in Behçet disease. Retina. 2012;32(7):1395–1402.
  • Park J, Cheon JH, Park Y, et al. Efficacy and tolerability of methotrexate therapy for refractory intestinal Behçet’s disease: a single center experience. Intest Res. 2018;16(2):315–318.
  • Chande N, Wang Y, MacDonald JK, et al. Methotrexate for induction of remission in ulcerative colitis. Cochrane Database Syst Rev. 2014;2014(8):CD006618.
  • Wang W, Zhou H, Liu L. Side effects of methotrexate therapy for rheumatoid arthritis: a systematic review. Eur J Med Chem. 2018;158:502–516.
  • Yang SL, Zhao FY, Song H, et al. Methotrexate associated renal impairment is related to delayed elimination of high-dose methotrexate. Scientific World J. 2015;2015:751703.
  • Feldkamp M, Carey JC. Clinical teratology counseling and consultation case report: low dose methotrexate exposure in the early weeks of pregnancy. Teratology. 1993;47(6):533–539.
  • Ransom JT. Mechanism of action of mycophenolate mofetil. Ther Drug Monit. 1995;17(6):681–684.
  • Suthanthiran M, Strom TB. Immunoregulatory drugs: mechanistic basis for use in organ transplantation. Pediatr Nephrol. 1997;11(5):651–657.
  • Neri P, Mariotti C, Cimino L, et al. Long-term control of cystoid macular oedema in noninfectious uveitis with mycophenolate mofetil. Int Ophthalmol. 2009;29(3):127–133.
  • Adler YD, Mansmann U, Zouboulis CC. Mycophenolate mofetil is ineffective in the treatment of mucocutaneous Adamantiades-Behçet’s disease. Dermatology. 2001;203(4):322–324.
  • Allison AC, Eugui EM. Mycophenolate mofetil and its mechanisms of action. Immunopharmacology. 2000;47(2–3):85–118.
  • Köse O, Simşek I, Pay S. Mycophenolate sodium in the treatment of mucocutaneous Behcet’s diseases. Int J Dermatol. 2011;50(7):895–896.
  • Shugaiv E, Tüzün E, Mutlu M, et al. Mycophenolate mofetil as a novel immunosuppressant in the treatment of neuro-Behçet’s disease with parenchymal involvement: presentation of four cases. Clin Exp Rheumatol. 2011;29(4 Suppl 67):S64–S67.
  • Al Maimouni H, Gladman DD, Ibañez D, et al. Switching treatment between mycophenolate mofetil and azathioprine in lupus patients: indications and outcomes. Arthritis Care Res (Hoboken). 2014;66(12):1905–1909.
  • Constantinescu S, Pai A, Coscia LA, et al. Breast-feeding after transplantation. Best Pract Res Clin Obstet Gynaecol. 2014;28(8):1163–1173.
  • Pillinger MH, Abramson SB. The neutrophil in rheumatoid arthritis. Rheum Dis Clin North Am. 1995;21(3):691–714.
  • Park YE, Cheon JH. Updated treatment strategies for intestinal Behçet’s disease. Korean J Intern Med. 2018;33(1):1–19.
  • Amos RS, Pullar T, Bax DE, et al. Sulphasalazine for rheumatoid arthritis: toxicity in 774 patients monitored for one to 11 years. Br Med J (Clin Res Ed). 1986;293(6544):420–423.
  • Plosker GL, Croom KF. Sulfasalazine: a review of its use in the management of rheumatoid arthritis. Drugs. 2005;6513:1825–1849. [published correction appears in Drugs. 2005;65(18):2591].
  • Lian BS, Busmanis I, Lee HY. Relapsing course of sulfasalazine-induced drug reaction with eosinophilia and systemic symptoms (DRESS) complicated by alopecia universalis and vitiligo. Ann Acad Med Singapore. 2018;47(11):492–493.
  • Houman MH, Hamzaoui K. Promising new therapies for Behcet’s disease. Eur J Intern Med. 2006;17(3):163–169.
  • Jung YS, Hong SP, Kim TI, et al. Long-term clinical outcomes and factors predictive of relapse after 5-aminosalicylate or sulfasalazine therapy in patients with intestinal Behçet disease. J Clin Gastroenterol. 2012;46(5):e38–e45.
  • Yilmaz S, Karadag O, Yazisiz V, et al. Systemic involvements and preferred treatments in a large cohort of Behçet’s disease. Rheumatol Int. 2013;33(12):3025–3030.
  • Ideguchi H, Suda A, Takeno M, et al. Gastrointestinal manifestations of Behçet’s disease in Japan: a study of 43 patients. Rheumatol Int. 2014;34(6):851–856.
  • Çirkinoğlu MS, Demir S, Bilginer Y, et al. Behçet’s disease in children: single-center experience. Turk Pediatri Ars. 2019;54(3):179–184.
  • Mogadam M, Dobbins WO 3rd, Korelitz BI, et al. Pregnancy in inflammatory bowel disease: effect of sulfasalazine and corticosteroids on fetal outcome. Gastroenterology. 1981;80(1):72–76.
  • Zao J, Koren G, Bozzo P. Using nitrofurantoin while breastfeeding a newborn. Can Fam Physician. 2014;60(6):539–540.
  • Mochizuki M, Masuda K, Sakane T, et al. A clinical trial of FK506 in refractory uveitis. Am J Ophthalmol. 1993;115(6):763‐769.
  • Li Z, Sun F, Zhang Y, et al. Tacrolimus induces insulin resistance and increases the glucose absorption in the jejunum: a potential mechanism of the diabetogenic effects. PLoS One. 2015;10(11):e0143405.
  • Kainz A, Harabacz I, Cowlrick IS, et al. Review of the course and outcome of 100 pregnancies in 84 women treated with tacrolimus. Transplantation. 2000;70(12):1718–1721.
  • Hiramatsu Y, Yoshida S, Kotani T, et al. Changes in the blood level, efficacy, and safety of tacrolimus in pregnancy and the lactation period in patients with systemic lupus erythematosus. Lupus. 2018;27(14):2245–2252.
  • Keifer JA, Guttridge DC, Ashburner BP, et al. Inhibition of NF-kappa B activity by thalidomide through suppression of IkappaB kinase activity. J Biol Chem. 2001;276(25):22382–22387.
  • Geitz H, Handt S, Zwingenberger K. Thalidomide selectively modulates the density of cell surface molecules involved in the adhesion cascade. Immunopharmacology. 1996;31(2–3):213–221.
  • Hamuryudan V, Mat C, Saip S, et al. Thalidomide in the treatment of the mucocutaneous lesions of the Behçet syndrome. A randomized, double-blind, placebo-controlled trial. Ann Intern Med. 1998;128(6):443–450.
  • Sharma NL, Sharma VC, Mahajan VK, et al. Thalidomide: an experience in therapeutic outcome and adverse reactions. J Dermatolog Treat. 2007;18(6):335–340.
  • Gardner-Medwin JM, Smith NJ, Powell RJ. Clinical experience with thalidomide in the management of severe oral and genital ulceration in conditions such as Behçet’s disease: use of neurophysiological studies to detect thalidomide neuropathy. Ann Rheum Dis. 1994;53(12):828–832.
  • Barbui T, Falanga A. Thalidomide and thrombosis in multiple myeloma. J Thromb Haemost. 2003;1(3):421–422.
  • Kural-Seyahi E, Fresko I, Seyahi N, et al. The long-term mortality and morbidity of Behçet syndrome: a 2-decade outcome survey of 387 patients followed at a dedicated center. Medicine (Baltimore). 2003;82(1):60–76.
  • Becatti M, Emmi G, Silvestri E, et al. Neutrophil activation promotes fibrinogen oxidation and thrombus formation in Behçet disease. Circulation. 2016;133(3):302–311.
  • Silingardi M, Iotti M, Trenti C, et al. Thalidomide, deep venous thrombosis and vasculitis. J Thromb Haemost. 2004;2(11):2062–2063.
  • Hatemi I, Hatemi G, Pamuk ON, et al. TNF-alpha antagonists and thalidomide for the management of gastrointestinal Behçet’s syndrome refractory to the conventional treatment modalities: a case series and review of the literature. Clin Exp Rheumatol. 2015;33(6 Suppl 94):S129–S137.
  • Radomsky CL, Levine N. Thalidomide. Dermatol Clin. 2001;19(1):87–103.
  • Thalidomide teratogenic effects linked to degradation of SALL4. after 60 years, researchers have now shed light on the mechanism underlying thalidomide’s devastating teratogenic effects. Am J Med Genet A. 2018;176(12): 2538–2539.
  • Turan B, Pfister K, Diener PA, et al. Soluble tumour necrosis factor receptors sTNFR1 and sTNFR2 are produced at sites of inflammation and are markers of arthritis activity in Behçet’s disease. Scand J Rheumatol. 2008;37(2):135–141.
  • Fabiani C, Vitale A, Emmi G, et al. Efficacy and safety of adalimumab in Behçet’s disease-related uveitis: a multicenter retrospective observational study. Clin Rheumatol. 2017;36(1):183–189.
  • Tosi GM, Sota J, Vitale A, et al. Efficacy and safety of certolizumab pegol and golimumab in the treatment of non-infectious uveitis. Clin Exp Rheumatol. 2019;37(4):680–683.
  • Agnholt J, Kaltoft K. Infliximab downregulates interferon-gamma production in activated gut T-lymphocytes from patients with Crohn’s disease. Cytokine. 2001;15(4):212–222.
  • Lügering A, Schmidt M, Lügering N, et al. Infliximab induces apoptosis in monocytes from patients with chronic active Crohn’s disease by using a caspase-dependent pathway. Gastroenterology. 2001;121(5):1145–1157.
  • van den Brande J, Hommes DW, Peppelenbosch MP. Infliximab induced T lymphocyte apoptosis in Crohn’s disease. J Rheumatol Suppl. 2005;74:26–30.
  • Bruns H, Meinken C, Schauenberg P, et al. Anti-TNF immunotherapy reduces CD8+ T cell-mediated antimicrobial activity against Mycobacterium tuberculosis in humans. J Clin Invest. 2009;119(5):1167–1177.
  • Wallis RS. Tumour necrosis factor antagonists: structure, function, and tuberculosis risks. Lancet Infect Dis. 2008;8(10):601–611.
  • Scallon B, Cai A, Solowski N, et al. Binding and functional comparisons of two types of tumor necrosis factor antagonists. J Pharmacol Exp Ther. 2002;301(2):418–426.
  • Ganem D, Prince AM. Hepatitis B virus infection–natural history and clinical consequences. N Engl J Med. 2004;350(11):1118–1129.
  • Giannitti C, Lopalco G, Vitale A, et al. Long-term safety of anti-TNF agents on the liver of patients with spondyloarthritis and potential occult hepatitis B viral infection: an observational multicentre study. Clin Exp Rheumatol. 2017;35(1):93–97.
  • Chung ES, Packer M, Lo KH, et al. Anti-TNF Therapy Against Congestive Heart Failure Investigators. Randomized double-blind placebo-controlled pilot trial of infliximab, a chimeric monoclonal antibody to tumor necrosis factor-alpha, in patients with moderate-to-severe heart failure: results of the anti-TNF therapy against congestive heart failure (ATTACH) trial. Circulation. 2003;107(25):3133–3140.
  • Pegoretti V, Baron W, Laman JD, et al. Selective modulation of TNF-TNFRs signaling: insights for multiple sclerosis treatment. Front Immunol. 2018;9:925.
  • Mariette X, Matucci-Cerinic M, Pavelka K, et al. Malignancies associated with tumour necrosis factor inhibitors in registries and prospective observational studies: a systematic review and meta-analysis. Ann Rheum Dis. 2011;70(11):1895–1904.
  • Melikoglu M, Fresko I, Mat C, et al. Short-term trial of etanercept in Behçet’s disease: a double blind, placebo controlled study. J Rheumatol. 2005;32(1):98–105.
  • Korzenik J, Larsen MD, Nielsen J, et al. Increased risk of developing Crohn’s disease or ulcerative colitis in 17 018 patients while under treatment with anti-TNF-α agents, particularly etanercept, for autoimmune diseases other than inflammatory bowel disease. Aliment Pharmacol Ther. 2019;50(3):289–294.
  • Toussirot É, Houvenagel É, Goëb V, et al. Development of inflammatory bowel disease during anti-TNF-α therapy for inflammatory rheumatic disease: a nationwide series. Joint Bone Spine. 2012;79(5):457–463.
  • Guignard S, Gossec L, Salliot C, et al. Efficacy of tumour necrosis factor blockers in reducing uveitis flares in patients with spondylarthropathy: a retrospective study. Ann Rheum Dis. 2006;65(12):1631–1634.
  • Lim LL, Fraunfelder FW, Rosenbaum JT. Do tumor necrosis factor inhibitors cause uveitis? A registry-based study. Arthritis Rheum. 2007;56(10):3248–3252.
  • Wendling D, Paccou J, Berthelot JM, et al. New onset of uveitis during anti-tumor necrosis factor treatment for rheumatic diseases. Semin Arthritis Rheum. 2011;41(3):503–510.
  • Raffeiner B, Ometto F, Bernardi L, et al. Inefficacy or paradoxical effect? Uveitis in ankylosing spondylitis treated with etanercept. Case Rep Med. 2014;2014:471319.
  • Arida A, Fragiadaki K, Giavri E, et al. Anti-TNF agents for Behçet’s disease: analysis of published data on 369 patients. Semin Arthritis Rheum. 2011;41(1):61–70.
  • Vallet H, Riviere S, Sanna A, et al. Efficacy of anti-TNF alpha in severe and/or refractory Behçet’s disease: multicenter study of 124 patients. J Autoimmun. 2015;62:67–74.
  • Tanida S, Inoue N, Kobayashi K, et al. Adalimumab for the treatment of Japanese patients with intestinal Behçet’s disease. Clin Gastroenterol Hepatol. 2015;13(5):940–8.e3.
  • Inoue N, Kobayashi K, Naganuma M, et al. Long-term safety and efficacy of adalimumab for intestinal Behçet’s disease in the open label study following a phase 3 clinical trial. Intest Res. 2017;15(3):395–401.
  • Vitale A, Emmi G, Lopalco G, et al. Adalimumab effectiveness in Behçet’s disease: short and long-term data from a multicenter retrospective observational study. Clin Rheumatol. 2017;36(2):451–455.
  • Fabiani C, Sota J, Vitale A, et al. Cumulative retention rate of adalimumab in patients with Behçet’s disease-related uveitis: a four-year follow-up study. Br J Ophthalmol. 2018;102(5):637–641.
  • Hassard PV, Binder SW, Nelson V, et al. Anti-tumor necrosis factor monoclonal antibody therapy for gastrointestinal Behçet’s disease: a case report. Gastroenterology. 2001;120(4):995–999.
  • Tugal-Tutkun I, Mudun A, Urgancioglu M, et al. Efficacy of infliximab in the treatment of uveitis that is resistant to treatment with the combination of azathioprine, cyclosporine, and corticosteroids in Behçet’s disease: an open-label trial. Arthritis Rheum. 2005;52(8):2478–2484.
  • Mastroianni A, Minutilli E, Mussi A, et al. Cytokine profiles during infliximab monotherapy in psoriatic arthritis. Br J Dermatol. 2005;153(3):531–536.
  • Wu LC, Zarrin AA. The production and regulation of IgE by the immune system. Nat Rev Immunol. 2014;14(4):247–259.
  • Finkelman FD, Khodoun MV, Strait R. Human IgE-independent systemic anaphylaxis. J Allergy Clin Immunol. 2016;137(6):1674–1680.
  • Maggi E, Vultaggio A, Matucci A. Acute infusion reactions induced by monoclonal antibody therapy. Expert Rev Clin Immunol. 2011;7(1):55–63.
  • Iwata S, Saito K, Yamaoka K, et al. Effects of anti-TNF-alpha antibody infliximab in refractory entero-Behçet’s disease. Rheumatology (Oxford). 2009;48(8):1012–1013.
  • Hibi T, Hirohata S, Kikuchi H, et al. Infliximab therapy for intestinal, neurological, and vascular involvement in Behçet disease: efficacy, safety, and pharmacokinetics in a multicenter, prospective, open-label, single-arm phase 3 study. Medicine (Baltimore). 2016;95(24):e3863. [published correction appears in Medicine (Baltimore). 2016;95(31):e5074].
  • Fabiani C, Sota J, Vitale A, et al. Ten-year retention rate of infliximab in patients with Behçet’s disease-related uveitis. Ocul Immunol Inflamm. 2019;27(1):34–39.
  • Cantini F, Niccoli L, Nannini C, et al. Rapid loss of efficacy of biosimilar infliximab in three patients with Behçet’s disease after switching from infliximab originator. Eur J Rheumatol. 2017;4(4):288–290.
  • Lopalco G, Venerito V, Cantarini L, et al. Long-term effectiveness and safety of switching from originator to biosimilar infliximab in patients with Behçet’s disease. Intern Emerg Med. 2019;14(5):719–722.
  • Mesquida M, Victoria Hernández M, Llorenç V, et al. Behçet disease-associated uveitis successfully treated with golimumab. Ocul Immunol Inflamm. 2013;21(2):160–162.
  • Stübgen JP. Tumor necrosis factor-alpha antagonists and neuropathy. Muscle Nerve. 2008;37(3):281–292.
  • Belluzzo M, Dozzo M, Pigatto E, et al. A case of neuromyotonia in Behçet disease during TNF-α antagonist therapy. Neurol Sci. 2014;35(10):1637–1639.
  • Santos-Gómez M, Calvo-Río V, Blanco R, et al. The effect of biologic therapy different from infliximab or adalimumab in patients with refractory uveitis due to Behçet’s disease: results of a multicentre open-label study. Clin Exp Rheumatol. 2016;34(6 Suppl 102):S34–S40.
  • Vitale A, Emmi G, Lopalco G, et al. Long-term efficacy and safety of golimumab in the treatment of multirefractory Behçet’s disease. Clin Rheumatol. 2017;36(9):2063–2069. [published correction appears in Clin Rheumatol. 2018 Sep 22].
  • Fabiani C, Sota J, Rigante D, et al. Rapid and sustained efficacy of golimumab in the treatment of multirefractory uveitis associated with Behçet’s disease. Ocul Immunol Inflamm. 2019;27(1):58–63.
  • Lopalco G, Emmi G, Gentileschi S, et al. Certolizumab pegol treatment in Behçet’s disease with different organ involvement: a multicenter retrospective observational study. Mod Rheumatol. 2017;27(6):1031–1035.
  • van Schouwenburg PA, Rispens T, Wolbink GJ. Immunogenicity of anti-TNF biologic therapies for rheumatoid arthritis. Nat Rev Rheumatol. 2013;9(3):164–172.
  • Mariette X, Förger F, Abraham B, et al. Lack of placental transfer of certolizumab pegol during pregnancy: results from CRIB, a prospective, postmarketing, pharmacokinetic study. Ann Rheum Dis. 2018;77(2):228–233.
  • Cantarini L, Lopalco G, Caso F, et al. Effectiveness and tuberculosis-related safety profile of interleukin-1 blocking agents in the management of Behçet’s disease. Autoimmun Rev. 2015;14(1):1–9.
  • Hamzaoui K, Hamza M, Ayed K. Production of TNF-alpha and IL-1 in active Behçet’s disease. J Rheumatol. 1990;17(10):1428–1429.
  • Pay S, Erdem H, Pekel A, et al. Synovial proinflammatory cytokines and their correlation with matrix metalloproteinase-3 expression in Behçet’s disease. Does interleukin-1beta play a major role in Behçet’s synovitis? Rheumatol Int. 2006;26(7):608–613.
  • Akman A, Ekinci NC, Kacaroglu H, et al. Relationship between periodontal findings and specific polymorphisms of interleukin-1alpha and −1beta in Turkish patients with Behçet’s disease. Arch Dermatol Res. 2008;300(1):19–26.
  • Castrichini M, Lazzerini PE, Gamberucci A, et al. The purinergic P2×7 receptor is expressed on monocytes in Behçet’s disease and is modulated by TNF-α. Eur J Immunol. 2014;44(1):227–238.
  • Lopalco G, Rigante D, Cantarini L, et al. The autoinflammatory side of recurrent pericarditis: enlightening the pathogenesis for a more rational treatment. Trends Cardiovasc Med. 2020;S1050-1738(20):30060–30068.
  • Sfikakis PP, Markomichelakis N, Alpsoy E, et al. Anti-TNF therapy in the management of Behcet’s disease–review and basis for recommendations. Rheumatology (Oxford). 2007;46(5):736–741.
  • Rigante D, Lopalco G, Vitale A, et al. Untangling the web of systemic autoinflammatory diseases. Mediators Inflamm. 2014;2014:948154.
  • Bettiol A, Lopalco G, Emmi G, et al. Unveiling the efficacy, safety, and tolerability of anti-interleukin-1 treatment in monogenic and multifactorial autoinflammatory diseases. Int J Mol Sci. 2019;20(8):1898.
  • Sota J, Vitale A, Insalaco A, et al. Safety profile of the interleukin-1 inhibitors anakinra and canakinumab in real-life clinical practice: a nationwide multicenter retrospective observational study. Clin Rheumatol. 2018;37(8):2233–2240.
  • Vitale A, Insalaco A, Sfriso P, et al. A snapshot on the on-label and off-label use of the interleukin-1 inhibitors in Italy among rheumatologists and pediatric rheumatologists: a nationwide multi-center retrospective observational study. Front Pharmacol. 2016;7:380.
  • Botsios C, Sfriso P, Furlan A, et al. Resistant Behçet disease responsive to anakinra. Ann Intern Med. 2008;149(4):284–286.
  • Cantarini L, Vitale A, Scalini P, et al. Anakinra treatment in drug resistant Behçet’s disease: a case series. Clin Rheumatol. 2015;34(7):1293–1301.
  • Emmi G, Talarico R, Lopalco G, et al. Efficacy and safety profile of anti-interleukin-1 treatment in Behçet’s disease: a multicenter retrospective study. Clin Rheumatol. 2016;35(5):1281–1286.
  • Grayson PC, Yazici Y, Merideth M, et al. Treatment of mucocutaneous manifestations in Behçet’s disease with anakinra: a pilot open-label study. Arthritis Res Ther. 2017;19(1):69.
  • Fabiani C, Vitale A, Emmi G, et al. Interleukin (IL)-1 inhibition with anakinra and canakinumab in Behçet’s disease-related uveitis: a multicenter retrospective observational study. Clin Rheumatol. 2017;36(1):191–197.
  • Fabiani C, Vitale A, Rigante D, et al. The presence of uveitis is associated with a sustained response to the interleukin (IL)-1 inhibitors anakinra and canakinumab in Behçet’s disease. Ocul Immunol Inflamm. 2020;28(2):298–304.
  • Tugal-Tutkun I, Pavesio C, De Cordoue A, et al. Use of gevokizumab in patients with Behçet’s disease uveitis: an international, randomized, double-masked, placebo-controlled study and open-label extension study. Ocul Immunol Inflamm. 2018;26(7):1023–1033.
  • Vitale A, Rigante D, Caso F, et al. Inhibition of interleukin-1 by canakinumab as a successful mono-drug strategy for the treatment of refractory Behçet’s disease: a case series. Dermatology. 2014;228(3):211–214.
  • Götestam Skorpen C, Hoeltzenbein M, Tincani A, et al. The EULAR points to consider for use of antirheumatic drugs before pregnancy, and during pregnancy and lactation. Ann Rheum Dis. 2016;75(5):795–810.
  • Youngstein T, Hoffmann P, Gul A, et al. International multi-centre study of pregnancy outcomes with interleukin-1 inhibitors. Rheumatology (Oxford). 2017;56:2102–2108.
  • Nanke Y, Yago T, Kotake S. The role of Th17 cells in the pathogenesis of Behçet’s disease. J Clin Med. 2017;6(7):74.
  • Di Scala G, Bettiol A, Cojan RD, et al. Efficacy of the anti-IL 17 secukinumab in refractory Behçet’s syndrome: a preliminary study. J Autoimmun. 2019;97:108–113.
  • Verstockt B, Deleenheer B, Van Assche G, et al. A safety assessment of biological therapies targeting the IL-23/IL-17 axis in inflammatory bowel diseases. Expert Opin Drug Saf. 2017;16(7):809–821.
  • Hueber W, Sands BE, Lewitzky S, et al. Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn’s disease: unexpected results of a randomised, double-blind placebo-controlled trial. Gut. 2012;61(12):1693–1700.
  • Neurath MF. Cytokines in inflammatory bowel disease. Nat Rev Immunol. 2014;14(5):329–342.
  • Dick AD, Tugal-Tutkun I, Foster S, et al. Secukinumab in the treatment of noninfectious uveitis: results of three randomized, controlled clinical trials. Ophthalmology. 2013;120(4):777–787.
  • Dincses E, Yurttas B, Esatoglu SN, et al. Secukinumab induced Behçet’s syndrome: a report of two cases. Oxf Med Case Reports. 2019;2019(5):omz041.
  • Fagni F, Bettiol A, Talarico R, et al. Long-term effectiveness and safety of secukinumab for treatment of refractory mucosal and articular Behçet’s phenotype: a multicentre study. Ann Rheum Dis. 2020;annrheumdis-2020-217108. Epub ahead of print. DOI:https://doi.org/10.1136/annrheumdis-2020-217108.
  • Salmaninejad A, Zamani MR, Shabgah AG, et al. Behçet’s disease: an immunogenetic perspective. J Cell Physiol. 2019;234(6):8055‐8074.
  • Lopalco G, Fabiani C, Venerito V, et al. Ustekinumab efficacy and safety in mucocutaneous multi-refractory Behçet’s disease. Clin Exp Rheumatol. 2017;35 Suppl 108(6):130–131.
  • Mirouse A, Barete S, Monfort JB, et al. Ustekinumab for Behçet’s disease. J Autoimmun. 2017;82:41–46.
  • Mirouse A, Barete S, Desbois AC, et al. Long-term outcome of ustekinumab therapy for Behçet’s disease. Arthritis Rheumatol. 2019;71(10):1727–1732.
  • Ekşioglu-Demiralp E, Kibaroglu A, Direskeneli H, et al. Phenotypic characteristics of B cells in Behçet’s disease: increased activity in B cell subsets. J Rheumatol. 1999;26(4):826–832.
  • van der Houwen TB, van Hagen PM, Timmermans WM, et al. Chronic signs of memory B cell activation in patients with Behçet’s disease are partially restored by anti-tumour necrosis factor treatment. Rheumatology (Oxford). 2017;56(1):134–144.
  • Davatchi F, Shams H, Rezaipoor M, et al. Rituximab in intractable ocular lesions of Behcet’s disease; randomized single-blind control study (pilot study). Int J Rheum Dis. 2010;13(3):246–252.
  • Shan D, Ledbetter JA, Press OW. Apoptosis of malignant human B cells by ligation of CD20 with monoclonal antibodies. Blood. 1998;91(5):1644–1652.
  • Shimabukuro-Vornhagen A, Gödel P, Subklewe M, et al. Cytokine release syndrome. J Immunother Cancer. 2018;6(1):56.
  • Sadreddini S, Noshad H, Molaeefard M, et al. Treatment of retinal vasculitis in Behçet’s disease with rituximab. Mod Rheumatol. 2008;18(3):306–308.
  • Messina MJ, Rodegher M, Scotti R, et al. Treatment of myelitis in Behçet’s disease with rituximab. BMJ Case Rep. 2014;2014:bcr2014204366.
  • Zhao BH, Oswald AE. Improved clinical control of a challenging case of Behçet’s disease with rituximab therapy. Clin Rheumatol. 2014;33(1):149–150.
  • Pelegrin L, Jakob E, Schmidt-Bacher A, et al. Experiences with rituximab for the treatment of autoimmune diseases with ocular involvement. J Rheumatol. 2014 Jan;41(1):84–90.
  • Santos-Gómez M, Calvo-Río V, Blanco R, et al. The effect of biologic therapy different from infliximab or adalimumab in patients with refractory uveitis due to Behçet’s disease: results of a multicentre open-label study. Clin Exp Rheumatol. 2016;Sep-Oct;34(6 Suppl 102):S34–S40.
  • Berger JR, Malik V, Lacey S, et al. Progressive multifocal leukoencephalopathy in rituximab-treated rheumatic diseases: a rare event. J Neurovirol. 2018 Jun;24(3):323–331.
  • Caldarelli-Stefano R, Vago L, Omodeo-Zorini E, et al. Detection and typing of JC virus in autopsy brains and extraneural organs of AIDS patients and non-immunocompromised individuals. J Neurovirol. 1999;5(2):125–133.
  • Agarwal S, Patrick J, Jones J, et al. Progressive multifocal leucoencephalopathy with Behçet’s disease: an insight into pathophysiology. Rheumatology (Oxford). 2017;56(4):668–670.
  • Stasi R, Del Poeta G, Stipa E, et al. Response to B-cell depleting therapy with rituximab reverts the abnormalities of T-cell subsets in patients with idiopathic thrombocytopenic purpura. Blood. 2007;110(8):2924–2930.
  • Barone M, Notarnicola A, Lopalco G, et al. Safety of long-term biologic therapy in rheumatologic patients with a previously resolved hepatitis B viral infection. Hepatology. 2015;62(1):40–46.
  • Chakravarty EF, Murray ER, Kelman A, et al. Pregnancy outcomes after maternal exposure to rituximab. Blood. 2011;117(5):1499–1506.
  • Bragnes Y, Boshuizen R, de Vries A, et al. Low level of Rituximab in human breast milk in a patient treated during lactation. Rheumatology (Oxford). 2017;56(6):1047–1048.
  • Borhani Haghighi A, Ittehadi H, Nikseresht AR, et al. CSF levels of cytokines in neuro-Behçet’s disease. Clin Neurol Neurosurg. 2009;111(6):507–510.
  • Hirohata S, Isshi K, Oguchi H, et al. Cerebrospinal fluid interleukin-6 in progressive Neuro-Behçet’s syndrome. Clin Immunol Immunopathol. 1997;82(1):12–17.
  • Ilbay A, Erden A, Sari A, et al. Successful treatment of amyloid A-type amyloidosis due to Behçet disease with tocilizumab. J Clin Rheumatol. 2019;25(4):43–45.
  • Dinoia L, Lopalco G, Cantarini L, et al. Long-term tocilizumab efficacy in a patient with psoriatic arthritis and AA amyloidosis. Clin Exp Rheumatol. 2017;35(1):170–171.
  • Kishimoto T. Interleukin-6: discovery of a pleiotropic cytokine. Arthritis Res Ther. 2006;8 Suppl 2(Suppl2):S2.
  • Garbers C, Aparicio-Siegmund S, Rose-John S. The IL-6/gp130/STAT3 signaling axis: recent advances towards specific inhibition. Curr Opin Immunol. 2015;34:75–82.
  • Shapiro LS, Farrell J, Borhani Haghighi A. Tocilizumab treatment for neuro-Behcet’s disease, the first report. Clin Neurol Neurosurg. 2012;114(3):297–298.
  • Urbaniak P, Hasler P, Kretzschmar S. Refractory neuro-Behçet treated by tocilizumab: a case report. Clin Exp Rheumatol. 2012;30(3 Suppl 72):S73–S75.
  • Addimanda O, Pipitone N, Pazzola G, et al. Tocilizumab for severe refractory neuro-Behçet: three cases IL-6 blockade in neuro-Behçet. Semin Arthritis Rheum. 2015;44(4):472–475.
  • Cantarini L, Lopalco G, Vitale A, et al. Paradoxical mucocutaneous flare in a case of Behçet’s disease treated with tocilizumab. Clin Rheumatol. 2015;34(6):1141–1143.
  • Lin ZQ, Kondo T, Ishida Y, et al. Essential involvement of IL-6 in the skin wound-healing process as evidenced by delayed wound healing in IL-6-deficient mice. J Leukoc Biol. 2003;73(6):713–721.
  • Lopalco G, Fabiani C, Sota J, et al. IL-6 blockade in the management of non-infectious uveitis. Clin Rheumatol. 2017;36(7):1459–1469.
  • Atienza-Mateo B, Calvo-Río V, Beltrán E, et al. Anti-interleukin 6 receptor tocilizumab in refractory uveitis associated with Behçet’s disease: multicentre retrospective study. Rheumatology (Oxford). 2018;57(5):856–864.
  • Ding Y, Li C, Liu J, et al. Tocilizumab in the treatment of severe and/or refractory vasculo-Behçet’s disease: a single-centre experience in China. Rheumatology (Oxford). 2018;57(11):2057–2059.
  • Hoffman E, Rahat MA, Feld J, et al. Effects of tocilizumab, an anti-interleukin-6 receptor antibody, on serum lipid and adipokine levels in patients with rheumatoid arthritis. Int J Mol Sci. 2019;20(18):4633.
  • Cacciapaglia F, Anelli MG, Rinaldi A, et al. Lipids and atherogenic indices fluctuation in rheumatoid arthritis patients on long-term tocilizumab treatment. Mediators Inflamm. 2018;2018:2453265.
  • Guglani L, Khader SA. Th17 cytokines in mucosal immunity and inflammation. Curr Opin HIV AIDS. 2010;5(2):120–127.
  • Schafer P. Apremilast mechanism of action and application to psoriasis and psoriatic arthritis. Biochem Pharmacol. 2012;83(12):1583–1590.
  • Hatemi G, Melikoglu M, Tunc R, et al. Apremilast for Behçet’s syndrome-a phase 2, placebo-controlled study. N Engl J Med. 2015;372(16):1510–1518.
  • Hatemi G, Mahr A, Ishigatsubo Y, et al. Trial of apremilast for oral ulcers in Behçet’s syndrome. N Engl J Med. 2019;381(20):1918–1928.
  • Epstein RS, Cummings NA, Sherwood EB, et al. Psychiatric aspects of Behçet’s syndrome. J Psychosom Res. 1970;14(2):161–172.
  • Deeks ED. Apremilast: A review in oral ulcers of Behçet’s disease. Drugs. 2020;80(2):181–188.
  • Lopalco G, Venerito V, Leccese P, et al. Real-world effectiveness of apremilast in multirefractory mucosal involvement of Behçet’s disease. Ann Rheum Dis. 2019;78(12):1736–1737.
  • De Luca G, Cariddi A, Campochiaro C, et al. Efficacy and safety of apremilast for Behçet’s syndrome: a real-life single-centre Italian experience. Rheumatology (Oxford). 2020;59(1):171–175.
  • Lambert JA, Raju SV, Tang LP, et al. Cystic fibrosis transmembrane conductance regulator activation by roflumilast contributes to therapeutic benefit in chronic bronchitis. Am J Respir Cell Mol Biol. 2014;50(3):549–558.
  • Langley A, Beecker J. Management of common side effects of apremilast. J Cutan Med Surg. 2018;22(4):415–421.
  • Abraham B, Sellin JH. Drug-induced diarrhea. Curr Gastroenterol Rep. 2007;9(5):365–372.
  • Atienza-Mateo B, Martin-Varillas JL, Loricera J, et al. Apremilast in refractory oral and/or genital ulcers in Behçet’s disease. Multicenter study of 49 cases in clinical practice. Ann Rheum Dis. 2019;78(Suppl 2):1182–1183.
  • Touzot M, Cacoub P, Bodaghi B, et al. IFN-α induces IL-10 production and tilt the balance between Th1 and Th17 in Behçet disease. Autoimmun Rev. 2015;14(5):370–375.
  • Tsambaos D, Eichelberg D, Goos M. Behçet’s syndrome: treatment with recombinant leukocyte alpha-interferon. Arch Dermatol Res. 1986;278(4):335–336.
  • Boyvat A, Sişman-Solak C, Gürler A. Long-term effects of interferon alpha 2A treatment in Behçet’s disease. Dermatology. 2000;201(1):40–43.
  • Zouboulis CC, Orfanos CE. Treatment of adamantiades-Behçet disease with systemic interferon alpha. Arch Dermatol. 1998;134(8):1010–1016.
  • Souvignet C, Lejeune O, Trepo C. Interferon-based treatment of chronic hepatitis C. Biochimie. 2007;89(6–7):894–898.
  • Alpsoy E, Durusoy C, Yilmaz E, et al. Interferon alfa-2a in the treatment of Behçet disease: a randomized placebo-controlled and double-blind study. Arch Dermatol. 2002;138(4):467–471.
  • Kötter I, Günaydin I, Zierhut M, et al. The use of interferon alpha in Behçet disease: review of the literature. Semin Arthritis Rheum. 2004;33(5):320–335.
  • Hamuryudan V, Ozyazgan Y, Fresko Y, et al. Interferon alfa combined with azathioprine for the uveitis of Behçet’s disease: an open study. Isr Med Assoc J. 2002;4(11 Suppl):928–930.
  • Kötter I, Hamuryudan V, Oztürk ZE, et al. Interferon therapy in rheumatic diseases: state-of-the-art 2010. Curr Opin Rheumatol. 2010;22(3):278–283.
  • Aydinoglu-Candan Ö, Araz-Erşan B, Gul A, et al. Anti-interferon alpha antibodies and autoantibodies in patients with Behçet’s disease uveitis treated with recombinant human interferon alpha-2a. Graefes Arch Clin Exp Ophthalmol. 2015;253(3):457–465.
  • Tomer Y, Blackard JT, Akeno N. Interferon alpha treatment and thyroid dysfunction. Endocrinol Metab Clin North Am. 2007;36(4):1051–1066.
  • Mandac JC, Chaudhry S, Sherman KE, et al. The clinical and physiological spectrum of interferon-alpha induced thyroiditis: toward a new classification. Hepatology. 2006;43(4):661–672.
  • Tran HA, Jones TL, Ianna EA, et al. Thyroid disease in chronic hepatitis C infection treated with combination interferon-α and ribavirin: management strategies and future perspective. Endocr Pract. 2013;19(2):292–300.
  • Yalçindag N, Köse HC. Comparison of the treatment results for Behçet uveitis in patients treated with infliximab and interferon. Ocul Immunol Inflamm. 2020;28(2):305–314.
  • Calgüneri M, Oztürk MA, Ertenli I, et al. Effects of interferon alpha treatment on the clinical course of refractory Behçet’s disease: an open study. Ann Rheum Dis. 2003;62(5):492–493.
  • Bolek EC, Sari A, Kilic L, et al. Interferon alpha might be an alternative therapeutic choice for refractory Neuro-Behçet’s disease. Mult Scler Relat Disord. 2019;29:153.
  • Balsat M, Etienne M, Elhamri M, et al. Successful pregnancies in patients with BCR-ABL-positive leukemias treated with interferon-alpha therapy during the tyrosine kinase inhibitors era. Eur J Haematol. 2018;101(6):774–780.
  • Lopalco G, Rigante D, Venerito V, et al. Update on the medical management of gastrointestinal Behçet’s disease. Mediators Inflamm. 2017;2017:1460491.
  • De Rosa G, Pardeo M, Rigante D. Current recommendations for the pharmacologic therapy in Kawasaki syndrome and management of its cardiovascular complications. Eur Rev Med Pharmacol Sci. 2007;11(5):301–308.
  • Hoffmann JHO, Enk AH. High-dose intravenous immunoglobulin in skin autoimmune disease. Front Immunol. 2019;10:1090.
  • Seider N, Beiran I, Scharf J, et al. Intravenous immunoglobulin therapy for resistant ocular Behçet’s disease. Br J Ophthalmol. 2001;85(11):1287–1288.
  • Rigante D, Valentini P, Rizzo D, et al. Responsiveness to intravenous immunoglobulins and occurrence of coronary artery abnormalities in a single-center cohort of Italian patients with Kawasaki syndrome. Rheumatol Int. 2010;30(6):841–846.
  • Guo Y, Tian X, Wang X, et al. Adverse effects of immunoglobulin therapy. Front Immunol. 2018;9:1299.
  • Rewald E, Jaksic JC. Behçet’s syndrome treated with high-dose intravenous IgG and low-dose aspirin. J R Soc Med. 1990;83(10):652–653.
  • Cantarini L, Stromillo ML, Vitale A, et al. Efficacy and safety of intravenous immunoglobulin treatment in refractory Behçet’s disease with different organ involvement: a case series. Isr Med Assoc J. 2016;18(3–4):238–242.
  • Perricone R, De Carolis C, Kröegler B, et al. Intravenous immunoglobulin therapy in pregnant patients affected with systemic lupus erythematosus and recurrent spontaneous abortion. Rheumatology (Oxford). 2008;47(5):646–651.
  • Rhen T, Cidlowski JA. Anti-inflammatory action of glucocorticoids-new mechanisms for old drugs. N Engl J Med. 2005;353(16):1711–1723.
  • Rigante D, Frediani B, Galeazzi M, et al. From the Mediterranean to the sea of Japan: the transcontinental odyssey of autoinflammatory diseases. Biomed Res Int. 2013;2013:485103.
  • Rigante D. A systematic approach to autoinflammatory syndromes: a spelling booklet for the beginner. Expert Rev Clin Immunol. 2017;13(6):571–597.
  • Rigante D. The broad-ranging panorama of systemic autoinflammatory disorders with specific focus on acute painful symptoms and hematologic manifestations in children. Mediterr J Hematol Infect Dis. 2018;10(1):e2018067.
  • Rigante D. Phenotype variability of autoinflammatory disorders in the pediatric patient: a pictorial overview. J Evid Based Med. 2020 Jul 6; Epub ahead of print. DOI:https://doi.org/10.1111/jebm.12406
  • Emmi G, Vitale A, Silvestri E, et al. Adalimumab-based treatment versus disease-modifying antirheumatic drugs for venous thrombosis in Behçet’s syndrome: a retrospective study of seventy patients with vascular involvement. Arthritis Rheumatol. 2018;70(9):1500–1507.
  • Emmi G, Bettiol A, Silvestri E, et al. Vascular Behçet’s syndrome: an update. Intern Emerg Med. 2019;14(5):645–652.
  • Alibaz-Oner F, Karadeniz A, Ylmaz S, et al. Behçet disease with vascular involvement: effects of different therapeutic regimens on the incidence of new relapses. Medicine (Baltimore). 2015;94(6):e494.
  • Tascilar K, Melikoglu M, Ugurlu S, et al. Vascular involvement in Behçet’s syndrome: a retrospective analysis of associations and the time course. Rheumatology. 2014;53:2018–2022.
  • Crowley J, Thaçi D, Joly P, et al. Long-term safety and tolerability of apremilast in patients with psoriasis: pooled safety analysis for ≥156 weeks from 2 phase 3, randomized, controlled trials (ESTEEM 1 and 2). J Am Acad Dermatol. 2017;77(2):310–317.e1.
  • Ahn HS, Lee D, Lee SY, et al. Increased cardiovascular risk and all-cause death in patients with Behçet disease: a Korean nationwide population-based dynamic cohort study. J Rheumatol. 2020;47(6):903–908.
  • Apalla Z, Psarakis E, Lallas A, et al. Psoriasis in patients with active lung cancer: is apremilast a safe option? Dermatol Pract Concept. 2019;9(4):300–301.
  • Nishi K, Luo H, Ishikura S, et al. Apremilast induces apoptosis of human colorectal cancer cells with mutant KRAS. Anticancer Res. 2017;37(7):3833–3839.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.