HHE Rheumatology and musculoskeletal supplement 2018 | Page 17

response and 15% with no response respectively. 13 GCA patients typically exceed a cumulative dose of 5000mg prednisolone over several years. 14 In a large UK retrospective study the average cumulative prednisolone use over the first two years from diagnosis was 8600mg; however, 33.4% received over 10,000mg and 3.3% more than 25,000mg. 2 It is estimated that for every 1000mg cumulative increase in GC dose, the adverse event hazard ratio increases by 3%. 14 Unfortunately, this does not treat the underlying inflammation and contributes to a higher incidence of steroid related side effects, estimated to affect 85% of patients with LV-GCA. 15 These side effects include diabetes, glaucoma, cataracts, hypertension, heart failure, osteoporosis, mood disturbance and increased susceptibility to infection, but this is Vascular US is a promising and rapidly developing area in GCA diagnosis, potentially avoiding the need for further invasive investigations such as temporal artery biopsy not by any means an exhaustive list. 2 GCA cohorts are particularly vulnerable due to their older age and higher prevalence of comorbidities. 16 EULAR taskforce recommendations suggest the risk of harm is low for the patients at long-term dosages of ≤5mg prednisone equivalent per day, whereas at dosages of >10mg/day, the risk of harm is elevated. 17 Between 5 and 10mg, the risk of harm is dependent on additional patient-specific factors. Not only are patients with relapsing and refractory GCA at higher risk of adverse effects secondary to toxic levels of glucocorticoids, but by definition the disease is sub-optimally controlled. This can lead directly to vascular damage such as aneurysms, dissection, rupture and stenotic disease. As such, the clinical and economic burden of these sub-groups is amplified. There remains a large unmet need for effective glucocorticoid-sparing agents. Currently there is no good evid ence for cDMARDs in GCA. One meta- analysis based on three small randomised-controlled trials (RCTs) suggested a role for methotrexate. 18 References 1 Patil P et al. Fast Track Pathway reduces sight loss in giant cell arteritis: results of a longitudinal observational cohort study. Clin Exp Rheumatol 2015;33(Suppl 89):S-103-6. 2 Petri H et al. Incidence of giant cell arteritis and characteristics of patients: Data-driven analysis of comorbidities. Arthritis Care Res 2015;67(3):390–5. 3 Dejaco C et al. The spectrum of giant cell arteritis and polymyalgia rheumatica: revisiting the concept go the disease. Rheumatology (Oxford) 2017;56:506–15. 4 Kermani T, Dasgupta B. Current and emerging therapies in large- vessel vasculitis. Rheumatology (Oxford) 2017;Oct 24 [Epub ahead of print]. 5 Dejaco C et al. EULAR recommendations for the use of imaging in large vessel vasculitis in clinical practice. Ann Rheum Dis 2018;0:1–8. 6 Chrysidis S et al. Ultrasound definitions for cranial and large vessel giant cell arteritis: Results of a reliability exercise on images and videos of the OMERACT Ultrasound Large Vessel Vasculitis Task Force. Arthritis Rheumatol 2016;68(Suppl 10). 7 Schmidt WA et al. Ultrasound of proximal upper extremity arteries to increase the diagnostic yield in large-vessel giant cell arteritis. Rheumatology (Oxford) 2008;47:96–101. 8 Schäfer VS et al. Ultrasound cut-off values for intimate-media thickness of temporal facial and axillary arteries in giant cell arteritis. Rheumatology (Oxford) 2017;56:1479–83. However its addition did not help to significantly reduce cumulative glucocorticoid dose or morbidity and mortality. 15 There are case series recommending the use of leflunomide and mycofenolate mofetil, but these are yet to be tested in an RCT setting. 15 Trials of biologic agents in GCA have had varying success. To date, no role has been found for use of anti-TNF agents, with infliximab, etanercept and adalimumab showing inefficacy. 15 Tocilizumab, an IL-6 receptor blocker, now has a good evidence base. IL-6-driven inflammation has been implicated in GCA since first described by Dasgupta and Panayi in 1990; however, it was not until recently that an agent utilising this pathway was available in GCA. 19 In the Phase III trial, GiACTA, 119 newly diagnosed and 132 relapsing patients were randomised to receive either weekly or fortnightly subcutaneous tocilizumab with a 26-week prednisone GC taper, versus the placebo arms with a 26- and 52-week prednisone taper alone. 20 Sustained prednisone-free remission was achieved in 56% and 53% of the weekly and fortnightly tocilizumab groups, respectively, compared with only 14% and 18% in the 26-week and 52-week placebo arms, respectively. Importantly, the cumulative prednisone dose was significantly lower in the treatment arm. Based on these results, tocilizumab has now been licensed for use by the US FDA and NICE in the UK. However, even in GiACTA, there were patients who did not achieve remission on tocilizumab, and it is not certain about its long-term impact on vascular damage. Other trials using biologics such as abatacept are currently ongoing, and we await the results. Ultimately there is still more work to be done in finding alternative treatments. Conclusions Introduction of ‘Fast Track Pathways’ can offer a prompt and secure diagnosis, reducing morbidity in patients with GCA and minimising inappropriate glucocorticoid use in those who do not. Vascular US is a promising and rapidly developing area in GCA and LV-GCA diagnosis, potentially avoiding the need for further invasive investigations such as TAB. There is a large unmet need beyond glucocorticoids in GCA and LV-GCA. There are some new promising treatments, including biologic therapies such as tocilizumab, but this will not be suitable for every patient. Further robust RCTs of other glucocorticoid-sparing agents are required. 9 Schmidt WA. Ultrasound in the diagnosis and management of giant cell arteritis. Rheumatology (Oxford) 2018;57:ii22–ii31. 10 Pipitone N, Versari A, Salvarani C. Role of imaging studies in the diagnosis and follow-up of large-vessel vasculitis: an update. Rheumatology (Oxford) 2008;47(4):403–8. 11 Blockmans D, Thorsten B, Schmidt W. Imaging for large-vessel vasculitis. Curr Opin Rheumatol 2009;21(1):19–28. 12 Tuckwell K et al. Newly diagnosed versus relapsing giant cell arteritis: Baseline data from the GiACTA trial. Semin Arthritis Rheum 2016;46(5):657–64. 13 Dasgupta B et al. 2012 provisional classification criteria for polymyalgia rheumatica: a European League Against Rhematism/American College 17 HHE 2018 | hospitalhealthcare.com of Rheumatology collaborative initiative. Ann Rheum Dis 2012;71(4):484–92. 14 Broder MS et al. Corticosteroid-related adverse events in patients with giant cell arteritis: A claims-based analysis. Semin Arthritis Rheum 2016;46(2):246–52. 15 Dejaco C et al. Giant cell arteritis and polymyalgia: current challenges and opportunities. Nat Rev Rheumatol 2017;13(10):578–92. 16 Proven A et al. Glucocorticoid therapy in giant cell arteritis: duration and adverse outcomes. Arthritis Rheum 2003;49(5): 703–8. 17 Strehl C et al. Defining conditions where long-term glucocorticoid treatment has an acceptably low level of harm to facilitate implementation of existing recommendations: viewpoints from an EULAR task force. Ann Rheum Dis 2016;75(6):952–7. 18 Mahr AD et al. Adjunctive methotrexate for treatment of giant cell arteritis: an individual patient data meta-analysis. Arthritis Rheum 2007;56(8):2789–97. 19 Dasgupta B, Panayi GS. Interlukin-6 in serum of patient with polymyalgia rheumatic and giant cell arteritis. Br J Rheumatol 1990;29:456–8. 20 Stone JH et al. Trial of tocilizumab in giant-cell arteritis. N Engl J Med 2017;377(4): 317–28.