361
Views
5
CrossRef citations to date
0
Altmetric
Review

Investigational drugs in systemic vasculitis

, , , , &
Pages 1049-1061 | Received 13 Jun 2017, Accepted 27 Jul 2017, Published online: 04 Aug 2017

References

  • Jennette JC, Falk RJ, Bacon PA, et al. 2012 revised International Chapel Hill Consensus Conference Nomenclature of Vasculitides. Arthritis Rheum. 2013 Jan;65(1):1–11.
  • Yates M, Watts RA, Bajema IM, et al. EULAR/ERA-EDTA recommendations for the management of ANCA-associated vasculitis. Ann Rheum Dis. 2016 Sep;75(9):1583–1594.
  • Xiao H, Schreiber A, Heeringa P, et al. Alternative complement pathway in the pathogenesis of disease mediated by anti-neutrophil cytoplasmic autoantibodies. Am J Pathol. 2007 Jan;170(1):52–64.
  • Wang H, Wang C, Zhao M-H, et al. Neutrophil extracellular traps can activate alternative complement pathways. Clin Exp Immunol. 2015 Sep;181(3):518–527.
  • Xing G, Chen M, Liu G, et al. Complement activation is involved in renal damage in human antineutrophil cytoplasmic autoantibody associated pauci-immune vasculitis. J Clin Immunol. 2009 May;29(3):282–291.
  • Gou S-J, Yuan J, Chen M, et al. Circulating complement activation in patients with anti-neutrophil cytoplasmic antibody-associated vasculitis. Kidney Int. 2013 Jan;83(1):129–137.
  • Hillmen P, Hall C, Marsh JCW, et al. Effect of eculizumab on hemolysis and transfusion requirements in patients with paroxysmal nocturnal hemoglobinuria. N Engl J Med. 2004 Feb 5;350(6):552–559.
  • Brodsky RA, Young NS, Antonioli E, et al. Multicenter phase 3 study of the complement inhibitor eculizumab for the treatment of patients with paroxysmal nocturnal hemoglobinuria. Blood. 2008 Feb 15;111(4):1840–1847.
  • Legendre CM, Licht C, Muus P, et al. Terminal complement inhibitor eculizumab in atypical hemolytic-uremic syndrome. N Engl J Med. 2013 Jun 6;368(23):2169–2181.
  • Xiao H, Dairaghi DJ, Powers JP, et al. C5a receptor (CD88) blockade protects against MPO-ANCA GN. J Am Soc Nephrol JASN. 2014 Feb;25(2):225–231.
  • Bekker P, Dairaghi D, Seitz L, et al. Characterization of pharmacologic and pharmacokinetic properties of CCX168, a potent and selective orally administered complement 5a receptor inhibitor, based on preclinical evaluation and randomized phase 1 clinical study. PLoS One. 2016;11(10):e0164646.
  • Luqmani RA, Bacon PA, Moots RJ, et al. Birmingham Vasculitis Activity Score (BVAS) in systemic necrotizing vasculitis. QJM Mon J Assoc Physicians. 1994 Nov;87(11):671–678.
  • Jayne DRW, Bruchfeld AN, Harper L, et al. Randomized trial of C5a receptor inhibitor avacopan in ANCA-associated vasculitis. J Am Soc Nephrol JASN. 2017 Apr 11.
  • Rasmussen N, Petersen J. Cellular immune responses and pathogenesis in c-ANCA positive vasculitides. J Autoimmun. 1993 Apr;6(2):227–236.
  • Lúdvíksson BR, Sneller MC, Chua KS, et al. Active Wegener’s granulomatosis is associated with HLA-DR+ CD4+ T cells exhibiting an unbalanced Th1-type T cell cytokine pattern: reversal with IL-10. J Immunol Baltim Md 1950. 1998 Apr 1;160(7):3602–3609.
  • Free ME, Bunch DO, McGregor JA, et al. Patients with antineutrophil cytoplasmic antibody-associated vasculitis have defective Treg cell function exacerbated by the presence of a suppression-resistant effector cell population. Arthritis Rheum. 2013 Jul;65(7):1922–1933.
  • Laffery KJ, Prowse SJ, Simeonovic CJ, et al. Immunobiology of tissue transplantation: a return to the passenger leukocyte concept. Annu Rev Immunol. 1983;1:143–173.
  • June CH, Bluestone JA, Nadler LM, et al. The B7 and CD28 receptor families. Immunol Today. 1994 Jul;15(7):321–331.
  • Walunas TL, Lenschow DJ, Bakker CY, et al. CTLA-4 can function as a negative regulator of T cell activation. Immunity. 1994 Aug;1(5):405–413.
  • Langford CA, Monach PA, Specks U, et al. An open-label trial of abatacept (CTLA4-IG) in non-severe relapsing granulomatosis with polyangiitis (Wegener’s). Ann Rheum Dis. 2014 Jul;73(7):1376–1379.
  • Takeuchi T, Iinuma H, Kunimoto S, et al. A new antitumor antibiotic, spergualin: isolation and antitumor activity. J Antibiot (Tokyo). 1981 Dec;34(12):1619–1621.
  • Nadeau K, Nadler SG, Saulnier M, et al. Quantitation of the interaction of the immunosuppressant deoxyspergualin and analogs with Hsc70 and Hsp90. Biochemistry (Mosc). 1994 Mar 8;33(9):2561–2567.
  • Sugawara A, Torigoe T, Tamura Y, et al. Polyamine compound deoxyspergualin inhibits heat shock protein-induced activation of immature dendritic cells. Cell Stress Chaperones. 2009 Mar;14(2):133–139.
  • Diegel ML, Nadler SG, Kiener PA. In vivo administration of 15-deoxyspergulin inhibits antigen-presenting cell stimulation of T cells and NF-kappaB activation. Int Immunopharmacol. 2002 Sep;2(10):1451–1464.
  • Perenyei M, Jayne DRW, Floßmann O. Gusperimus: immunological mechanism and clinical applications. Rheumatol Oxf Engl. 2014 Oct;53(10):1732–1741.
  • Holcombe H, Mellman I, Janeway CA, et al. The immunosuppressive agent 15-deoxyspergualin functions by inhibiting cell cycle progression and cytokine production following naive T cell activation. J Immunol Baltim Md 1950. 2002 Nov 1;169(9):4982–4989.
  • Borg AJ, Kumagai-Braesch M, Möller E. 15-Deoxyspergualin inhibits interleukin 6 production in in vitro stimulated human lymphocytes. Transpl Immunol. 1996 Jun;4(2):133–143.
  • Birck R, Newman M, Braun C, et al. 15-Deoxyspergualin and cyclophosphamide, but not mycophenolate mofetil, prolong survival and attenuate renal disease in a murine model of ANCA-associated crescentic nephritis. Nephrol Dial Transplant Off Publ Eur Dial Transpl Assoc Eur Ren Assoc. 2006 Jan;21(1):58–63.
  • Birck R, Warnatz K, Lorenz HM, et al. 15-Deoxyspergualin in patients with refractory ANCA-associated systemic vasculitis: a six-month open-label trial to evaluate safety and efficacy. J Am Soc Nephrol JASN. 2003 Feb;14(2):440–447.
  • Schmitt WH, Birck R, Heinzel PA, et al. Prolonged treatment of refractory Wegener’s granulomatosis with 15-deoxyspergualin: an open study in seven patients. Nephrol Dial Transplant Off Publ Eur Dial Transpl Assoc Eur Ren Assoc. 2005 Jun;20(6):1083–1092.
  • Flossmann O, Baslund B, Bruchfeld A, et al. Deoxyspergualin in relapsing and refractory Wegener’s granulomatosis. Ann Rheum Dis. 2009 Jul;68(7):1125–1130.
  • Walsh M, Chaudhry A, Jayne D. Long-term follow-up of relapsing/refractory anti-neutrophil cytoplasm antibody associated vasculitis treated with the lymphocyte depleting antibody alemtuzumab (CAMPATH-1H). Ann Rheum Dis. 2008 Sep;67(9):1322–1327.
  • Voswinkel J, Müller A, Lamprecht P. Is PR3-ANCA formation initiated in Wegener’s granulomatosis lesions? Granulomas as potential lymphoid tissue maintaining autoantibody production. Ann N Y Acad Sci. 2005 Jun;1051:12–19.
  • Krumbholz M, Specks U, Wick M, et al. BAFF is elevated in serum of patients with Wegener’s granulomatosis. J Autoimmun. 2005 Dec;25(4):298–302.
  • Cupps TR, Edgar LC, Fauci AS. Suppression of human B lymphocyte function by cyclophosphamide. J Immunol Baltim Md 1950. 1982 Jun;128(6):2453–2457.
  • Jones RB, Tervaert JWC, Hauser T, et al. Rituximab versus cyclophosphamide in ANCA-associated renal vasculitis. N Engl J Med. 2010 Jul 15;363(3):211–220.
  • Stone JH, Merkel PA, Spiera R, et al. Rituximab versus cyclophosphamide for ANCA-associated vasculitis. N Engl J Med. 2010 Jul 15;363(3):221–232.
  • Navarra SV, Guzmán RM, Gallacher AE, et al. Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: a randomised, placebo-controlled, phase 3 trial. Lancet Lond Engl. 2011 Feb 26;377(9767):721–731.
  • Comarmond C, Pagnoux C, Khellaf M, et al. Eosinophilic granulomatosis with polyangiitis (Churg-Strauss): clinical characteristics and long-term followup of the 383 patients enrolled in the French Vasculitis Study Group cohort. Arthritis Rheum. 2013 Jan;65(1):270–281.
  • Schnabel A, Csernok E, Braun J, et al. Inflammatory cells and cellular activation in the lower respiratory tract in Churg-Strauss syndrome. Thorax. 1999 Sep;54(9):771–778.
  • Hellmich B, Csernok E, Gross WL. Proinflammatory cytokines and autoimmunity in Churg-Strauss syndrome. Ann N Y Acad Sci. 2005 Jun;1051:121–131.
  • Noga O, Hanf G, Brachmann I, et al. Effect of omalizumab treatment on peripheral eosinophil and T-lymphocyte function in patients with allergic asthma. J Allergy Clin Immunol. 2006 Jun;117(6):1493–1499.
  • Djukanović R, Wilson SJ, Kraft M, et al. Effects of treatment with anti-immunoglobulin E antibody omalizumab on airway inflammation in allergic asthma. Am J Respir Crit Care Med. 2004 Sep 15;170(6):583–593.
  • Hart TK, Cook RM, Zia-Amirhosseini P, et al. Preclinical efficacy and safety of mepolizumab (SB-240563), a humanized monoclonal antibody to IL-5, in cynomolgus monkeys. J Allergy Clin Immunol. 2001 Aug;108(2):250–257.
  • Kahn J-E, Grandpeix-Guyodo C, Marroun I, et al. Sustained response to mepolizumab in refractory Churg-Strauss syndrome. J Allergy Clin Immunol. 2010 Jan;125(1):267–270.
  • Kim S, Marigowda G, Oren E, et al. Mepolizumab as a steroid-sparing treatment option in patients with Churg-Strauss syndrome. J Allergy Clin Immunol. 2010 Jun;125(6):1336–1343.
  • Wechsler ME, Akuthota P, Jayne D, et al. Mepolizumab or placebo for eosinophilic granulomatosis with polyangiitis. N Engl J Med. 2017 May 18;376(20):1921–1932.
  • Detoraki A, Di Capua L, Varricchi G, et al. Omalizumab in patients with eosinophilic granulomatosis with polyangiitis: a 36-month follow-up study. J Asthma Off J Assoc Care Asthma. 2016;53(2):201–206.
  • Brouet J-C, Clauvel J-P, Danon F, et al. Biologic and clinical significance of cryoglobulins. A report of 86 cases. Am J Med. 1974 Nov;57(5):775–788.
  • Muchtar E, Magen H, Gertz MA. How I treat cryoglobulinemia. Blood. 2017 Jan 19;129(3):289–298.
  • Smith KA. Interleukin-2: inception, impact, and implications. Science. 1988 May 27;240(4856):1169–1176.
  • Ahmadzadeh M, Rosenberg SA. IL-2 administration increases CD4+ CD25(hi) Foxp3+ regulatory T cells in cancer patients. Blood. 2006 Mar 15;107(6):2409–2414.
  • Boyer O, Saadoun D, Abriol J, et al. CD4+CD25+ regulatory T-cell deficiency in patients with hepatitis C-mixed cryoglobulinemia vasculitis. Blood. 2004 May 1;103(9):3428–3430.
  • Saadoun D, Rosenzwajg M, Joly F, et al. Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis. N Engl J Med. 2011 Dec 1;365(22):2067–2077.
  • Ito M, Murakami K, Suzuki T, et al. Enhanced expression of lymphomagenesis-related genes in peripheral blood B cells of chronic hepatitis C patients. Clin Immunol Orlando Fla. 2010 Jun;135(3):459–465.
  • Terrier B, Krastinova E, Marie I, et al. Management of noninfectious mixed cryoglobulinemia vasculitis: data from 242 cases included in the CryoVas survey. Blood. 2012 Jun 21;119(25):5996–6004.
  • Sneller MC, Hu Z, Langford CA. A randomized controlled trial of rituximab following failure of antiviral therapy for hepatitis C virus-associated cryoglobulinemic vasculitis. Arthritis Rheum. 2012 Mar;64(3):835–842.
  • Visentini M, Ludovisi S, Petrarca A, et al. A phase II, single-arm multicenter study of low-dose rituximab for refractory mixed cryoglobulinemia secondary to hepatitis C virus infection. Autoimmun Rev. 2011 Sep;10(11):714–719.
  • Visentini M, Tinelli C, Colantuono S, et al. Efficacy of low-dose rituximab for the treatment of mixed cryoglobulinemia vasculitis: phase II clinical trial and systematic review. Autoimmun Rev. 2015 Oct;14(10):889–896.
  • Roccatello D, Sciascia S, Rossi D, et al. The challenge of treating hepatitis C virus-associated cryoglobulinemic vasculitis in the era of anti-CD20 monoclonal antibodies and direct antiviral agents. Oncotarget. 2017 Apr 9.
  • Roccatello D, Sciascia S, Baldovino S, et al. Improved (4 plus 2) rituximab protocol for severe cases of mixed cryoglobulinemia: a 6-year observational study. Am J Nephrol. 2016;43(4):251–260.
  • Sakane T, Takeno M, Suzuki N, et al. Behçet’s disease. N Engl J Med. 1999 Oct 21;341(17):1284–1291.
  • Yazici H, Yurdakul S, Hamuryudan V. Behçet disease. Curr Opin Rheumatol. 2001 Jan;13(1):18–22.
  • Hatemi G, Silman A, Bang D, et al. Management of Behçet disease: a systematic literature review for the European League Against Rheumatism evidence-based recommendations for the management of Behçet disease. Ann Rheum Dis. 2009 Oct;68(10):1528–1534.
  • 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 Apr;26(4):826–832.
  • Hamzaoui K, Houman H, Ben Dhifallah I, et al. Serum BAFF levels and skin mRNA expression in patients with Behçet’s disease. Clin Exp Rheumatol. 2008 Aug;26(4 Suppl 50):S64–71.
  • Adam B, Calikoglu E. Serum interleukin-6, procalcitonin and C-reactive protein levels in subjects with active Behçet’s disease. J Eur Acad Dermatol Venereol JEADV. 2004 May;18(3):318–320.
  • 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 Aug;13(3):246–252.
  • Aktulga E, Altaç M, Müftüoglu A, et al. A double blind study of colchicine in Behçet’s disease. Haematologica. 1980 Jun;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 Nov;44(11):2686–2692.
  • Hidi R, Timmermans S, Liu E, et al. Phosphodiesterase and cyclic adenosine monophosphate-dependent inhibition of T-lymphocyte chemotaxis. Eur Respir J. 2000 Feb;15(2):342–349.
  • Kumar N, Goldminz AM, Kim N, et al. Phosphodiesterase 4-targeted treatments for autoimmune diseases. BMC Med. 2013 Apr 4;11:96.
  • Hatemi G, Melikoglu M, Tunc R, et al. Apremilast for Behçet’s syndrome – a phase 2, placebo-controlled study. N Engl J Med. 2015 Apr 16;372(16):1510–1518.
  • Gül A. Behçet’s disease as an autoinflammatory disorder. Curr Drug Targets Inflamm Allergy. 2005 Feb;4(1):81–83.
  • Dinarello CA. Interleukin-1beta and the autoinflammatory diseases. N Engl J Med. 2009 Jun 4;360(23):2467–2470.
  • Mege JL, Dilsen N, Sanguedolce V, et al. Overproduction of monocyte derived tumor necrosis factor alpha, interleukin (IL) 6, IL-8 and increased neutrophil superoxide generation in Behçet’s disease. A comparative study with familial Mediterranean fever and healthy subjects. J Rheumatol. 1993 Sep;20(9):1544–1549.
  • Zou J, Guan J-L. Interleukin-1-related genes polymorphisms in Turkish patients with Behçet disease: a meta-analysis. Mod Rheumatol. 2014 Mar;24(2):321–326.
  • Roell MK, Issafras H, Bauer RJ, et al. Kinetic approach to pathway attenuation using XOMA 052, a regulatory therapeutic antibody that modulates interleukin-1beta activity. J Biol Chem. 2010 Jul 2;285(27):20607–20614.
  • Gül A, Tugal-Tutkun I, Dinarello CA, et al. Interleukin-1β-regulating antibody XOMA 052 (gevokizumab) in the treatment of acute exacerbations of resistant uveitis of Behcet’s disease: an open-label pilot study. Ann Rheum Dis. 2012 Apr;71(4):563–566.
  • Tugal-Tutkun I, Kadayifcilar S, Khairallah M, et al. Safety and efficacy of gevokizumab in patients with Behçet’s disease uveitis: results of an exploratory phase 2 study. Ocul Immunol Inflamm. 2017 Feb;25(1):62–70.
  • Lachmann HJ, Kone-Paut I, Kuemmerle-Deschner JB, et al. Use of canakinumab in the cryopyrin-associated periodic syndrome. N Engl J Med. 2009 Jun 4;360(23):2416–2425.
  • 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 Jan;36(1):191–197.
  • 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. Dermatol Basel Switz. 2014;228(3):211–214.
  • 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 Mar 24;19(1):69.
  • 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 May;14(5):370–375.
  • Habibagahi Z, Habibagahi M, Heidari M. Raised concentration of soluble form of vascular endothelial cadherin and IL-23 in sera of patients with Behçet’s disease. Mod Rheumatol. 2010 Apr;20(2):154–159.
  • Gheita TA, Gamal SM, Shaker I, et al. Clinical significance of serum interleukin-23 and A/G gene (rs17375018) polymorphism in Behçets disease: relation to neuro-Behçet, uveitis and disease activity. Joint Bone Spine. 2015 May;82(3):213–215.
  • Kim J, Park JA, Lee EY, et al. Imbalance of Th17 to Th1 cells in Behçet’s disease. Clin Exp Rheumatol. 2010 Aug;28(4 Suppl 60):S16–19.
  • Remmers EF, Cosan F, Kirino Y, et al. Genome-wide association study identifies variants in the MHC class I, IL10, and IL23R-IL12RB2 regions associated with Behçet’s disease. Nat Genet. 2010 Aug;42(8):698–702.
  • Mizuki N, Meguro A, Ota M, et al. Genome-wide association studies identify IL23R-IL12RB2 and IL10 as Behçet’s disease susceptibility loci. Nat Genet. 2010 Aug;42(8):703–706.
  • Reddy M, Davis C, Wong J, et al. Modulation of CLA, IL-12R, CD40L, and IL-2Ralpha expression and inhibition of IL-12- and IL-23-induced cytokine secretion by CNTO 1275. Cell Immunol. 2007 May;247(1):1–11.
  • Mirouse A, Barete S, Monfort J-B, et al. Ustekinumab for Behçet’s disease. J Autoimmun. 2017 May 5;82:41–46.
  • Poulter LW, Lehner T. Immunohistology of oral lesions from patients with recurrent oral ulcers and Behçet’s syndrome. Clin Exp Immunol. 1989 Nov;78(2):189–195.
  • Gül A, Esin S, Dilsen N, et al. Immunohistology of skin pathergy reaction in Behçet’s disease. Br J Dermatol. 1995 Jun;132(6):901–907.
  • Mohammad AJ, Smith RM, Chow YW, et al. Alemtuzumab as remission induction therapy in Behçet disease: a 20-year experience. J Rheumatol. 2015 Oct;42(10):1906–1913.
  • Gonzalez-Gay MA, Miranda-Filloy JA, Lopez-Diaz MJ, et al. Giant cell arteritis in northwestern Spain: a 25-year epidemiologic study. Medicine (Baltimore). 2007 Mar;86(2):61–68.
  • Hachulla E, Boivin V, Pasturel-Michon U, et al. Prognostic factors and long-term evolution in a cohort of 133 patients with giant cell arteritis. Clin Exp Rheumatol. 2001 Apr;19(2):171–176.
  • Proven A, Gabriel SE, Orces C, et al. Glucocorticoid therapy in giant cell arteritis: duration and adverse outcomes. Arthritis Rheum. 2003 Oct 15;49(5):703–708.
  • Weyand CM, Hicok KC, Hunder GG, et al. Tissue cytokine patterns in patients with polymyalgia rheumatica and giant cell arteritis. Ann Intern Med. 1994 Oct 1;121(7):484–491.
  • Hernández-Rodríguez J, Segarra M, Vilardell C, et al. Tissue production of pro-inflammatory cytokines (IL-1beta, TNFalpha and IL-6) correlates with the intensity of the systemic inflammatory response and with corticosteroid requirements in giant-cell arteritis. Rheumatol Oxf Engl. 2004 Mar;43(3):294–301.
  • Hoffman GS, Cid MC, Rendt-Zagar KE, et al. Infliximab for maintenance of glucocorticosteroid-induced remission of giant cell arteritis: a randomized trial. Ann Intern Med. 2007 May 1;146(9):621–630.
  • Martínez-Taboada VM, Rodríguez-Valverde V, Carreño L, et al. A double-blind placebo controlled trial of etanercept in patients with giant cell arteritis and corticosteroid side effects. Ann Rheum Dis. 2008 May;67(5):625–630.
  • Seror R, Baron G, Hachulla E, et al. Adalimumab for steroid sparing in patients with giant-cell arteritis: results of a multicentre randomised controlled trial. Ann Rheum Dis. 2014 Dec;73(12):2074–2081.
  • Emilie D, Liozon E, Crevon MC, et al. Production of interleukin 6 by granulomas of giant cell arteritis. Hum Immunol. 1994 Jan;39(1):17–24.
  • van der Geest KSM, Abdulahad WH, Rutgers A, et al. Serum markers associated with disease activity in giant cell arteritis and polymyalgia rheumatica. Rheumatol Oxf Engl. 2015 Aug;54(8):1397–1402.
  • Mihara M, Kasutani K, Okazaki M, et al. Tocilizumab inhibits signal transduction mediated by both mIL-6R and sIL-6R, but not by the receptors of other members of IL-6 cytokine family. Int Immunopharmacol. 2005 Nov;5(12):1731–1740.
  • Seitz M, Reichenbach S, Bonel HM, et al. Rapid induction of remission in large vessel vasculitis by IL-6 blockade. A case series. Swiss Med Wkly. 2011 Jan;17(141):w13156.
  • Beyer C, Axmann R, Sahinbegovic E, et al. Anti-interleukin 6 receptor therapy as rescue treatment for giant cell arteritis. Ann Rheum Dis. 2011 Oct;70(10):1874–1875.
  • Unizony S, Arias-Urdaneta L, Miloslavsky E, et al. Tocilizumab for the treatment of large-vessel vasculitis (giant cell arteritis, Takayasu arteritis) and polymyalgia rheumatica. Arthritis Care Res. 2012 Nov;64(11):1720–1729.
  • Salvarani C, Magnani L, Catanoso M, et al. Tocilizumab: a novel therapy for patients with large-vessel vasculitis. Rheumatol Oxf Engl. 2012 Jan;51(1):151–156.
  • Loricera J, Blanco R, Hernández JL, et al. Tocilizumab in giant cell arteritis: multicenter open-label study of 22 patients. Semin Arthritis Rheum. 2015 Jun;44(6):717–723.
  • Villiger PM, Adler S, Kuchen S, et al. Tocilizumab for induction and maintenance of remission in giant cell arteritis: a phase 2, randomised, double-blind, placebo-controlled trial. Lancet Lond Engl. 2016 May 7;387(10031):1921–1927.
  • Xu Z, Bouman-Thio E, Comisar C, et al. Pharmacokinetics, pharmacodynamics and safety of a human anti-IL-6 monoclonal antibody (sirukumab) in healthy subjects in a first-in-human study. Br J Clin Pharmacol. 2011 Aug;72(2):270–281.
  • Smolen JS, Weinblatt ME, Sheng S, et al. Sirukumab, a human anti-interleukin-6 monoclonal antibody: a randomised, 2-part (proof-of-concept and dose-finding), phase II study in patients with active rheumatoid arthritis despite methotrexate therapy. Ann Rheum Dis. 2014 Sep;73(9):1616–1625.
  • Aletaha D, Bingham CO, Tanaka Y, et al. Efficacy and safety of sirukumab in patients with active rheumatoid arthritis refractory to anti-TNF therapy (SIRROUND-T): a randomised, double-blind, placebo-controlled, parallel-group, multinational, phase 3 study. Lancet Lond Engl. 2017 Mar 25;389(10075):1206–1217.
  • Rovin BH, van Vollenhoven RF, Aranow C, et al. A multicenter, randomized, double-blind, placebo-controlled study to evaluate the efficacy and safety of treatment with sirukumab (CNTO 136) in patients with active lupus nephritis. Arthritis Rheumatol Hoboken NJ. 2016 Sep;68(9):2174–2183.
  • Enjuanes A, Benavente Y, Hernández-Rodríguez J, et al. Association of NOS2 and potential effect of VEGF, IL6, CCL2 and IL1RN polymorphisms and haplotypes on susceptibility to GCA–a simultaneous study of 130 potentially functional SNPs in 14 candidate genes. Rheumatol Oxf Engl. 2012 May;51(5):841–851.
  • Ly K-H, Stirnemann J, Liozon E, et al. Interleukin-1 blockade in refractory giant cell arteritis. Jt Bone Spine Rev Rhum. 2014 Jan;81(1):76–78.
  • Deng J, Younge BR, Olshen RA, et al. Th17 and Th1 T-cell responses in giant cell arteritis. Circulation. 2010 Feb 23;121(7):906–915.
  • Espígol-Frigolé G, Corbera-Bellalta M, Planas-Rigol E, et al. Increased IL-17A expression in temporal artery lesions is a predictor of sustained response to glucocorticoid treatment in patients with giant-cell arteritis. Ann Rheum Dis. 2013 Sep 1;72(9):1481–1487.
  • Terrier B, Geri G, Chaara W, et al. Interleukin-21 modulates Th1 and Th17 responses in giant cell arteritis. Arthritis Rheum. 2012 Jun;64(6):2001–2011.
  • Márquez A, Hernández-Rodríguez J, Cid MC, et al. Influence of the IL17A locus in giant cell arteritis susceptibility. Ann Rheum Dis. 2014 Sep;73(9):1742–1745.
  • Shi JG, Chen X, Lee F, et al. The pharmacokinetics, pharmacodynamics, and safety of baricitinib, an oral JAK 1/2 inhibitor, in healthy volunteers. J Clin Pharmacol. 2014 Dec;54(12):1354–1361.
  • Keystone EC, Taylor PC, Drescher E, et al. Safety and efficacy of baricitinib at 24 weeks in patients with rheumatoid arthritis who have had an inadequate response to methotrexate. Ann Rheum Dis. 2015 Feb;74(2):333–340.
  • Papp KA, Menter MA, Raman M, et al. A randomized phase 2b trial of baricitinib, an oral Janus kinase (JAK) 1/JAK2 inhibitor, in patients with moderate-to-severe psoriasis. Br J Dermatol. 2016 Jun;174(6):1266–1276.
  • Genovese MC, Kremer J, Zamani O, et al. Baricitinib in patients with refractory rheumatoid arthritis. N Engl J Med. 2016 Mar 31;374(13):1243–1252.
  • Fleischmann R, Schiff M, van der Heijde D, et al. Baricitinib, methotrexate, or combination in patients with rheumatoid arthritis and no or limited prior disease-modifying antirheumatic drug treatment. Arthritis Rheumatol Hoboken NJ. 2017 Mar;69(3):506–517.
  • Smolen JS, Kremer JM, Gaich CL, et al. Patient-reported outcomes from a randomised phase III study of baricitinib in patients with rheumatoid arthritis and an inadequate response to biological agents (RA-BEACON). Ann Rheum Dis. 2016 Apr;76(4):694–700.
  • Conway R, O’Neill L, O’Flynn E, et al. Ustekinumab for the treatment of refractory giant cell arteritis. Ann Rheum Dis. 2016 Aug;75(8):1578–1579.
  • Brack A, Geisler A, Martinez-Taboada VM, et al. Giant cell vasculitis is a T cell-dependent disease. Mol Med Camb Mass. 1997 Aug;3(8):530–543.
  • Langford CA, Cuthbertson D, Ytterberg SR, et al. A randomized, double-blind trial of abatacept (CTLA-4Ig) for the treatment of giant cell arteritis. Arthritis Rheumatol. 2017 Apr 1;69(4):837–845.
  • Mwipatayi BP, Jeffery PC, Beningfield SJ, et al. Takayasu arteritis: clinical features and management: report of 272 cases. ANZ J Surg. 2005 Mar;75(3):110–117.
  • Maksimowicz-McKinnon K, Clark TM, Hoffman GS. Limitations of therapy and a guarded prognosis in an American cohort of Takayasu arteritis patients. Arthritis Rheum. 2007 Mar;56(3):1000–1009.
  • Hoffman GS, Leavitt RY, Kerr GS, et al. Treatment of glucocorticoid-resistant or relapsing Takayasu arteritis with methotrexate. Arthritis Rheum. 1994 Apr;37(4):578–582.
  • Seko Y. Takayasu arteritis: insights into immunopathology. Jpn Heart J. 2000 Jan;41(1):15–26.
  • Seko Y, Takahashi N, Tada Y, et al. Restricted usage of T-cell receptor Vgamma-Vdelta genes and expression of costimulatory molecules in Takayasu’s arteritis. Int J Cardiol. 2000 Aug 31;75(Suppl 1):S77-83-87.
  • Langford CA, Cuthbertson D, Ytterberg SR, et al. A randomized, double-blind trial of abatacept (CTLA-4Ig) for the treatment of Takayasu arteritis. Arthritis Rheumatol Hoboken NJ. 2017 Apr;69(4):846–853.
  • Noris M, Daina E, Gamba S, et al. Interleukin-6 and RANTES in Takayasu arteritis: a guide for therapeutic decisions? Circulation. 1999 Jul 6;100(1):55–60.
  • Park MC, Lee SW, Park YB, et al. Serum cytokine profiles and their correlations with disease activity in Takayasu’s arteritis. Rheumatol Oxf Engl. 2006 May;45(5):545–548.
  • Seko Y, Sato O, Takagi A, et al. Restricted usage of T-cell receptor Valpha-Vbeta genes in infiltrating cells in aortic tissue of patients with Takayasu’s arteritis. Circulation. 1996 May 15;93(10):1788–1790.
  • Nakaoka Y, Higuchi K, Arita Y, et al. Tocilizumab for the treatment of patients with refractory Takayasu arteritis. Int Heart J. 2013;54(6):405–411.
  • Abisror N, Mekinian A, Lavigne C, et al. Tocilizumab in refractory Takayasu arteritis: a case series and updated literature review. Autoimmun Rev. 2013 Oct;12(12):1143–1149.
  • Mekinian A, Comarmond C, Resche-Rigon M, et al. Efficacy of biological-targeted treatments in takayasu arteritis: multicenter, retrospective study of 49 patients. Circulation. 2015 Nov 3;132(18):1693–1700.
  • Loricera J, Blanco R, Hernández JL, et al. Tocilizumab in patients with Takayasu arteritis: a retrospective study and literature review. Clin Exp Rheumatol. 2016 Jun;34(3 Suppl 97):S44–53.
  • Sneller MC. Granuloma formation, implications for the pathogenesis of vasculitis. Cleve Clin J Med. 2002;69(Suppl 2):SII40–43.
  • Tripathy NK, Chauhan SK, Nityanand S. Cytokine mRNA repertoire of peripheral blood mononuclear cells in Takayasu’s arteritis. Clin Exp Immunol. 2004 Nov;138(2):369–374.
  • Tripathy NK, Gupta PC, Nityanand S. High TNF-alpha and low IL-2 producing T cells characterize active disease in Takayasu’s arteritis. Clin Immunol Orlando Fla. 2006 Mar;118(2–3):154–158.
  • Comarmond C, Plaisier E, Dahan K, et al. Anti TNF-α in refractory Takayasu’s arteritis: cases series and review of the literature. Autoimmun Rev. 2012 Jul;11(9):678–684.
  • Schmidt J, Kermani TA, Bacani AK, et al. Tumor necrosis factor inhibitors in patients with Takayasu arteritis: experience from a referral center with long-term followup. Arthritis Care Res. 2012 Jul;64(7):1079–1083.
  • Hoffman GS, Merkel PA, Brasington RD, et al. Anti-tumor necrosis factor therapy in patients with difficult to treat Takayasu arteritis. Arthritis Rheum. 2004 Jul;50(7):2296–2304.
  • Molloy ES, Langford CA, Clark TM, et al. Anti-tumour necrosis factor therapy in patients with refractory Takayasu arteritis: long-term follow-up. Ann Rheum Dis. 2008 Nov;67(11):1567–1569.

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.