174
Views
4
CrossRef citations to date
0
Altmetric
Review

Tailoring biologic therapy for real-world rheumatoid arthritis patients

ORCID Icon, , & ORCID Icon
Pages 661-674 | Received 02 Sep 2020, Accepted 03 Nov 2020, Published online: 15 Nov 2020

References

  • Smolen JS, Aletaha D, Barton A, et al. Rheumatoid arthritis. Nat Rev Dis Primers. 2018;4:18001.
  • Singh JA, Saag KG, Bridges SL Jr, et al. American College of Rheumatology guideline for the treatment of rheumatoid arthritis. Arthritis Rheumatol. 2016;68:1–26.
  • Smolen JS, Landewé R, Bijlsma J, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2019 update. Ann Rheum Dis. 2020;79:685–699.
  • Castañeda S, Atienza-Mateo B, Martín-Varillas JL, et al. Anakinra for the treatment of adult-onset Still’s disease. Expert Rev Clin Immunol. 2018;14:979–992.
  • Ogawa Y, Takahashi N, Kaneko A, et al. Association between seropositivity and discontinuation of tumor necrosis factor inhibitors due to ineffectiveness in rheumatoid arthritis. Clin Rheumatol. 2019;38:2757–2763.
  • Sokolove J, Schiff M, Fleischmann R, et al. Impact of baseline anti-cyclic citrullinated peptide-2 antibody concentration on efficacy outcomes following treatment with subcutaneous abatacept or adalimumab: 2-year results from the AMPLE trial. Ann Rheum Dis. 2016;75:709–714.
  • Isaacs JD, Cohen SB, Mery P, et al. Effect of baseline rheumatoid factor and anticitrullinated peptide antibody serotype on rituximab clinical response: a meta-analysis. Ann Rheum Dis. 2013;72:329–336.
  • Cappelli LC, Palmer JL, Kremer J, et al. 3rd. Tocilizumab treatment leads to improvement in disease activity regardless of CCP status in rheumatoid arthritis. Semin Arthritis Rheum. 2017;47:165–169.
  • Jansen DTSL, Emery P, Smolen JS, et al. Conversion to seronegative status after abatacept treatment in patients with early and poor prognostic rheumatoid arthritis is associated with better radiographic outcomes and sustained remission: post hoc analysis of the AGREE study. RMD Open. 2018;4:e000564.
  • Oryoji K, Yoshida K, Kashiwado Y, et al. Shared epitope positivity is related to efficacy of abatacept in rheumatoid arthritis. Ann Rheum Dis. 2018;77:1234–1236.
  • Couderc M, Mathieu S, Pereira B, et al. Predictive factors of rituximab response in rheumatoid arthritis: results from a French university hospital. Arthritis Care Res. 2013;65:648–652.
  • Chatzidionysiou K, Lie E, Nasanov E, et al. Highest clinical effectiveness of rituximab in autoantibody-positive patients with rheumatoid arthritis and in those for whom no more than one previous TNF antagonist has failed: pooled data from 10 European registries. Ann Rheum Dis. 2011;70:1575–1580.
  • Cohen SB, Emery P, Greenwald MW, et al. Rituximab for rheumatoid arthritis refractory to anti-tumor necrosis factor therapy: results of a multicenter randomized, double-blind, placebo-controlled, phase III trial evaluating primary efficacy and safety at twenty-four weeks. Arthritis Rheum. 2006;54:2793–2806.
  • Porter D, van Melckebeke J, Dale J, et al. Tumour necrosis factor inhibition versus rituximab for patients with rheumatoid arthritis who require biological treatment (ORBIT): an open-label, randomised controlled, non-inferiority, trial. Lancet. 2016;388:239–247.
  • Genovese MC, McKay JD, Nasonov EL, et al. Interleukin-6 receptor inhibition with tocilizumab reduces disease activity in rheumatoid arthritis with inadequate response to disease-modifying antirheumatic drugs: the tocilizumab in combination with traditional disease-modifying antirheumatic drug therapy study. Arthritis Rheum. 2008;58:2968–2980.
  • Smolen JS, Beaulieu A, Rubbert-Roth A, et al. Effect of interleukin-6 receptor inhibition with tocilizumab in patients with rheumatoid arthritis (OPTION study): a double-blind, placebo-controlled, randomised trial. Lancet. 2008;371:987–997.
  • Kremer JM, Blanco R, Brzosko M, et al. Tocilizumab inhibits structural joint damage in rheumatoid arthritis patients with inadequate responses to methotrexate: results from the double-blind treatment phase of a randomized placebo-controlled trial of tocilizumab safety and prevention of structural joint damage at one year. Arthritis Rheum. 2011;63:609–621.
  • Jones G, Sebba A, Gu J, et al. Comparison of tocilizumab monotherapy versus methotrexate monotherapy in patients with moderate to severe rheumatoid arthritis: the AMBITION study. Ann Rheum Dis. 2010;69:88–96.
  • Noack M, Miossec P. Selected cytokine pathways in rheumatoid arthritis. Semin Immunopathol. 2017;39:365–383.
  • Romano C, Del Mastro A, Sellitto A, et al. Tocilizumab reduces complement C3 and C4 serum levels in rheumatoid arthritis patients. Clin Rheumatol. 2018;37:1695–1700.
  • Gabay C, Emery P, van Vollenhoven R, et al. Tocilizumab monotherapy versus adalimumab monotherapy for treatment of rheumatoid arthritis (ADACTA): a randomised, double-blind, controlled phase 4 trial. Lancet. 2013;381:1541–1550.
  • Backhaus M, Kaufmann J, Richter C, et al. Comparison of tocilizumab and tumour necrosis factor inhibitors in rheumatoid arthritis: a retrospective analysis of 1603 patients managed in routine clinical practice. Clin Rheumatol. 2015;34:673–681.
  • Choy EH, Bernasconi C, Aassi M, et al. Treatment of rheumatoid arthritis with anti-tumor necrosis factor or tocilizumab therapy as first biologic agent in a global comparative observational study. Arthritis Care Res. 2017;69:1484–1494.
  • Bijlsma JWJ, Welsing PMJ, Woodworth TG, et al. Early rheumatoid arthritis treated with tocilizumab, methotrexate, or their combination (U-Act-Early): a multicentre, randomised, double-blind, double-dummy, strategy trial. Lancet. 2016;388:343–355.
  • Genovese MC, Fleischmann R, Kivitz AJ, et al. Sarilumab plus methotrexate in patients with active rheumatoid arthritis and inadequate response to methotrexate: results of a phase III study. Arthritis Rheumatol. 2015;67:1424–1437.
  • Fleischmann R, van Adelsberg J, Lin Y, et al. Sarilumab and nonbiologic disease-modifying antirheumatic drugs in patients with active rheumatoid arthritis and inadequate response or intolerance to tumor necrosis factor inhibitors. Arthritis Rheumatol. 2017;69:277–290.
  • Burmester GR, Lin Y, Patel R, et al. Efficacy and safety of sarilumab monotherapy versus adalimumab monotherapy for the treatment of patients with active rheumatoid arthritis (MONARCH): a randomised, double-blind, parallel-group phase III trial. Ann Rheum Dis. 2017;76:840–847.
  • Conway R, Low C, Coughlan RJ, et al. Methotrexate and lung disease in rheumatoid arthritis: a meta-analysis of randomized controlled trials. Arthritis Rheumatol. 2014;66:803–812.
  • Roubille C, Haraoui B. Interstitial lung diseases induced or exacerbated by DMARDS and biologic agents in rheumatoid arthritis: a systematic literature review. Semin Arthritis Rheum. 2014;43:613–626.
  • Panopoulos ST, Sfikakis PP. Biological treatments and connective tissue disease associated interstitial lung disease. Curr Opin Pulm Med. 2011;17:362–367.
  • Nakashita T, Ando K, Kaneko N, et al. Potential risk of TNF inhibitors on the progression of interstitial lung disease in patients with rheumatoid arthritis. BMJ Open. 2014;14:4.
  • Kawashiri SY, Kawakami A, Sakamoto N, et al. A fatal case of acute exacerbation of interstitial lung disease in a patient with rheumatoid arthritis during treatment with tocilizumab. Rheumatol Int. 2012;32:4023–4026.
  • Wendling D, Vidon C, Godfrin-Valnet M, et al. Exacerbation of combined pulmonary fibrosis and emphysema syndrome during tocilizumab therapy for rheumatoid arthritis. Joint Bone Spine. 2013;80:670–671.
  • Fernández-Díaz C, Loricera J, Castañeda S, et al. Abatacept in patients with rheumatoid arthritis and interstitial lung disease: A national multicenter study of 63 patients. Semin Arthritis Rheum. 2018;48:22–27.
  • Kurata I, Tsuboi H, Terasaki M, et al. Effect of biological disease-modifying anti-rheumatic drugs on airway and interstitial lung disease in patients with rheumatoid arthritis. Intern Med. 2019;58:1703–1712.
  • Curtis JR, Sarsour K, Napalkov P, et al. Incidence and complications of interstitial lung disease in users of tocilizumab, rituximab, abatacept and anti-tumor necrosis factor α agents, a retrospective cohort study. Arthritis Res Ther. 2015;17:319.
  • Fernández-Díaz C, Castañeda S, Melero-González RB, et al. Abatacept in interstitial lung disease associated with rheumatoid arthritis: national multicenter study of 263 patients. Rheumatology (Oxford). 2020. DOI:10.1093/rheumatology/keaa621.
  • Carrasco Cubero C, Chamizo Carmona E, Vela Casasempere P. Systematic review of the impact of drugs on diffuse interstitial lung disease associated with rheumatoid arthritis. Reumatol Clin. 2020. DOI:10.1016/j.reuma.2020.04.015
  • Vadillo C, Nieto MA, Romero-Bueno F, et al. Efficacy of rituximab in slowing down progression of rheumatoid arthritis-related interstitial lung disease: data from the NEREA registry. Rheumatology (Oxford). 2020;59:2099–2108.
  • Makino H, Yoshinaga Y, Yamasaki Y, et al. Renal involvement in rheumatoid arthritis: analysis of renal biopsy specimens from 100 patients. Mod Rheumatol. 2002;12:148–154.
  • Horii Y, Muraguchi A, Iwano M, et al. Involvement of IL-6 in mesangial proliferative glomerulonephritis. J Immunol. 1989;143:3949–3955.
  • Real de Asùa D, Costa R, Galvàn JM, et al. Systemic AA amyloidosis: epidemiology, diagnosis, and management. Clin Epidemiol. 2014;6:369–377.
  • Helin H, Korpela M, Mustonen J, et al. Renal biopsy findings and clinicopathologic correlations in rheumatoid arthritis. Arthritis Rheum. 1995;38:242–247.
  • Karstila K, Korpela M, Sihvonen S, et al. Prognosis of clinical renal disease and incidence of new renal findings in patients with rheumatoid arthritis: follow-up of a population-based study. Clin Rheumatol. 2007;26:2089–2095.
  • Nakamura T, Higashi S, Tomoda K, et al. Efficacy of etanercept in patients with AA amyloidosis secondary to rheumatoid arthritis. Clin Exp Rheumatol. 2007;25:518–522.
  • Esatoglu S, Hatemi G, Ugurlu S, et al. Long-term follow-up of secondary amyloidosis patients treated with tumor necrosis factor inhibitor therapy: a STROBE-compliant observational study. Medicine (Baltimore). 2017;96:e7859.
  • Lane T, Gillmore JD, Wechalekar AD, et al. Therapeutic blockade of interleukin-6 by tocilizumab in the management of AA amyloidosis and chronic inflammatory disorders: a case series and review of the literature. Clin Exp Rheumatol. 2015;33(6 Suppl 94):S46–53.
  • Okuda Y. AA amyloidosis - Benefits and prospects of IL-6 inhibitors. Mod Rheumatol. 2019;29:268–274.
  • Feng J, Chen Q, Yu F, et al. Body mass index and risk of rheumatoid arthritis: a meta-analysis of observational studies. Medicine (Baltimore). 2016;95:e2859.
  • Qin B, Yang M, Fu H, et al. Body mass index and the risk of rheumatoid arthritis: a systematic review and dose-response meta-analysis. Arthritis Res Ther. 2015;17:86.
  • Vidal C, Barnetche T, Morel J, et al. Association of body mass index categories with disease activity and radiographic joint damage in rheumatoid arthritis: a systematic review and metaanalysis. J Rheumatol. 2015;42:2261–2269.
  • Liu Y, Hazlewood GS, Kaplan GG, et al. Impact of obesity on remission and disease activity in rheumatoid arthritis: a systematic review and meta-analysis. Arthritis Care Res. 2017;69:157–165.
  • Gremese E, Carletto A, Padovan M, et al. Obesity and reduction of the response rate to anti-tumor necrosis factor α in rheumatoid arthritis: an approach to a personalized medicine. Arthritis Care Res. 2013;65:94–100.
  • Shan J, Zhang J. Impact of obesity on the efficacy of different biologic agents in inflammatory diseases: A systematic review and meta-analysis. Joint Bone Spine. 2019;86:173–183.
  • Ottaviani S, Gardette A, Roy C, et al. Body mass index and response to rituximab in rheumatoid arthritis. Joint Bone Spine. 2015;82:432–436.
  • Iannone F, Fanizzi R, Notarnicola A, et al. Obesity reduces the drug survival of second line biological drugs following a first TNF-α inhibitor in rheumatoid arthritis patients. Joint Bone Spine. 2015;82:187–191.
  • Liu W, Baker RD, Bhatia T, et al. Pathogenesis of nonalcoholic steatohepatitis. Cell Mol Life Sci. 2016;73:1969–1987.
  • Koca SS, Bahcecioglu IH, Poyrazoglu OK, et al. The treatment with antibody of TNF-alpha reduces the inflammation, necrosis and fibrosis in the non-alcoholic steatohepatitis induced by methionine- and choline-deficient diet. Inflammation. 2008;31:91–98.
  • Schramm C, Schneider A, Marx A, et al. Adalimumab could suppress the activity of non alcoholic steatohepatitis (NASH). Z Gastroenterol. 2008;46:1369–1371.
  • Mori S, Arima N, Ito M, et al. Non-alcoholic steatohepatitis-like pattern in liver biopsy of rheumatoid arthritis patients with persistent transaminitis during low-dose methotrexate treatment. PLoS One. 2018;13:e0203084.
  • Romano C, Esposito S, Ferrara R, et al. Choosing the most appropriate biologic therapy for Crohn’s disease according to concomitant extra-intestinal manifestations, comorbidities, or physiologic conditions. Expert Opin Biol Ther. 2020;20:49–62.
  • Rehman K, Akash MSH, Liaqat A, et al. Role of interleukin-6 in development of insulin resistance and type 2 diabetes mellitus. Crit Rev Eukaryot Gene Expr. 2017;27:229–236.
  • Solomon DH, Love TJ, Canning C, et al. Risk of diabetes among patients with rheumatoid arthritis, psoriatic arthritis and psoriasis. Ann Rheum Dis. 2010;69:2114–2117.
  • Chen DY, Chen YM, Hsieh TY, et al. Significant effects of biologic therapy on lipid profiles and insulin resistance in patients with rheumatoid arthritis. Arthritis Res Ther. 2015;17:52.
  • Schultz O, Oberhauser F, Saech J, et al. Effects of inhibition of interleukin-6 signalling on insulin sensitivity and lipoprotein (a) levels in human subjects with rheumatoid diseases. PLoS One. 2010;5:e14328.
  • Genovese MC, Burmester GR, Hagino O, et al. Effect of sarilumab on glycosylated hemoglobin in patients with rheumatoid arthritis and diabetes. Ann Rheum Dis. 2019;78(S2):1128.
  • Castañeda S, Remuzgo-Martínez S, López-Mejías R, et al. Rapid beneficial effect of the IL-6 receptor blockade on insulin resistance and insulin sensitivity in non-diabetic patients with rheumatoid arthritis. Clin Exp Rheumatol. 2019;37:465–473.
  • Orban T, Bundy B, Becker DJ, et al. Co-stimulation modulation with abatacept in patients with recent-onset type 1 diabetes: a randomised, double-blind, placebo-controlled trial. Lancet. 2011;378:412–419.
  • Orban T, Bundy B, Becker DJ, et al. Costimulation modulation with abatacept in patients with recent-onset type 1 diabetes: follow-up 1 year after cessation of treatment. Diabetes Care. 2014;37:1069–1075.
  • Pescovitz MD, Greenbaum CJ, Krause-Steinrauf H, et al. Rituximab, B-lymphocyte depletion, and preservation of beta-cell function. N Engl J Med. 2009;361:2143–2152.
  • Pescovitz MD, Greenbaum CJ, Bundy B, et al. B-lymphocyte depletion with rituximab and β-cell function: two-year results. Diabetes Care. 2014;37:453–459.
  • Artifoni M, Rothschild PR, Brézin A, et al. Ocular inflammatory diseases associated with rheumatoid arthritis. Nat Rev Rheumatol. 2014;10:108–116.
  • Akpek EK, Thorne JE, Qazi FA, et al. Evaluation of patients with scleritis for systemic disease. Ophthalmology. 2004;111:501–506.
  • Doctor P, Sultan A, Syed S, et al. Infliximab for the treatment of refractory scleritis. Br J Ophthalmol. 2010;94:579–583.
  • Murphy CC, Ayliffe WH, Booth A, et al. Tumor necrosis factor alpha blockade with infliximab for refractory uveitis and scleritis. Ophthalmology. 2004;111:352–356.
  • Atchia II, Kidd CE, Bell RW. Rheumatoid arthritis-associated necrotizing scleritis and peripheral ulcerative keratitis treated successfully with infliximab. J Clin Rheumatol. 2006;12:291–293.
  • Ashok D, Ayliffe WH, Kiely PD. Necrotizing scleritis associated with rheumatoid arthritis: long-term remission with high-dose infliximab therapy. Rheumatology (Oxford). 2005;44:950–951.
  • Chauhan S, Kamal A, Thompson RN, et al. Rituximab for treatment of scleritis associated with rheumatoid arthritis. Br J Ophthalmol. 2009;93:984–985.
  • Iaccheri B, Androudi S, Bocci EB, et al. Rituximab treatment for persistent scleritis associated with rheumatoid arthritis. Ocul Immunol Inflamm. 2010;18:223–225.
  • Smith JR, Levinson RD, Holland GN, et al. Differential efficacy of tumor necrosis factor inhibition in the management of inflammatory eye disease and associated rheumatic disease. Arthritis Rheum. 2001;45:252–257.
  • Thomas JW, Pflugfelder SC. Therapy of progressive rheumatoid arthritis-associated corneal ulceration with infliximab. Cornea. 2005;24:742–744.
  • Albert M, Beltrán E, Martínez-Costa L. Rituximab in rheumatoid arthritis-associated peripheral ulcerative keratitis. Arch Soc Esp Oftalmol. 2011;86:118–120.
  • Dominguez-Casas LC, Sánchez-Bilbao L, Calvo-Río V, et al. Biologic therapy in severe and refractory peripheral ulcerative keratitis (PUK). Multicenter study of 34 patients. Semin Arthritis Rheum. 2020;50:608–615.
  • Makol A, Matteson EL, Warrington KJ. Rheumatoid vasculitis: an update. Curr Opin Rheumatol. 2015;27:63–70.
  • Coffey CM, Richter MD, Crowson CS, et al. Rituximab therapy for systemic rheumatoid vasculitis: indications, outcomes and adverse events. J Rheumatol. 2020;47:518–523.
  • Puéchal X, Gottenberg JE, Berthelot JM, et al. Rituximab therapy for systemic vasculitis associated with rheumatoid arthritis: results from the AutoImmunity and Rituximab Registry. Arthritis Care Res (Hoboken). 2012;64:331–339.
  • Unger L, Kayser M, Nusslein HG. Successful treatment of severe rheumatoid vasculitis by infliximab. Ann Rheum Dis. 2003;62:587–588.
  • Puéchal X, Miceli-Richard C, Mejjad O, et al. Antitumour necrosis factor treatment in patients with refractory systemic vasculitis associated with rheumatoid arthritis. Ann Rheum Dis. 2008;67:880–884.
  • Ashida A, Murata H, Mikoshiba Y, et al. Successful treatment of rheumatoid vasculitis-associated skin ulcer with a TNF-α antagonist. Int J Dermatol. 2014;53:e154–156.
  • Jarrett SJ, Cunnane G, Conaghan PG, et al. Anti-tumor necrosis factor-alpha therapy-induced vasculitis: case series. J Rheumatol. 2003;30:2287–2291.
  • Mohan N, Edwards ET, Cupps TR, et al. Leukocytoclastic vasculitis associated with tumor necrosis factor-alpha blocking agents. J Rheumatol. 2004;31:1955–1958.
  • Sokumbi O, Wetter DA, Makol A, et al. Vasculitis associated with tumor necrosis factor-alpha inhibitors. Mayo Clin Proc. 2012;87:739–745.
  • Woody M, Warren D, Speck L, et al. Leukocytoclastic vasculitis drug reaction to certolizumab pegol. Proc Bayl Univ Med Cent. 2017;30:213–214.
  • Fujii W, Kohno M, Ishino H, et al. The rapid efficacy of abatacept in a patient with rheumatoid vasculitis. Mod Rheumatol. 2012;22:630–634.
  • Al Attar L, Shaver T. Abatacept as a therapeutic option for rheumatoid vasculitis. Cureus. 2018;10:e2506.
  • Sumida K, Ubara Y, Takemoto F, et al. Successful treatment with humanised anti-interleukin 6 receptor antibody for multidrug-refractory and antitumour necrosis factor-resistant systemic rheumatoid vasculitis. Clin Exp Rheumatol. 2011;29:S133.
  • Yoshida S, Takeuchi T, Sawaki H, et al. Successful treatment with tocilizumab of pericarditis associated with rheumatoid arthritis. Mod Rheumatol. 2014;24:677–680.
  • Iijima T, Suwabe T, Sumida K, et al. Tocilizumab improves systemic rheumatoid vasculitis with necrotizing crescentic glomerulonephritis. Mod Rheumatol. 2015;25:138–142.
  • Weinreb N, Rabinowitz A, Dellaripa PF. Beneficial response to rituximab in refractory Felty syndrome. J Clin Rheumatol. 2006;12:48.
  • Salama A, Schneider U, Dörner T. Beneficial response to rituximab in a patient with haemolysis and refractory Felty syndrome. Ann Rheum Dis. 2008;67:894–895.
  • Chandra PA, Margulis Y, Schiff C. Rituximab is useful in the treatment of Felty’s syndrome. Am J Ther. 2008;15:321–322.
  • Narváez J, Domingo-Domenech E, Gómez-Vaquero C, et al. Biological agents in the management of Felty’s syndrome: a systematic review. Semin Arthritis Rheum. 2012;41:658–668.
  • Sarp U, Ataman S. A beneficial long-term and consistent response to rituximab in the treatment of refractory neutropenia and arthritis in a patient with Felty syndrome. J Clin Rheumatol. 2014;20:398.
  • Ayzenberg M, Shenberger KN. Successful treatment of a large cutaneous ulcer and improvement in the hematologic manifestations of felty syndrome with rituximab. J Clin Rheumatol. 2014;20:440–441.
  • Pukšić S, Mitrović J, Morović-Vergles J. Rituximab: a safe treatment in a patient with refractory Felty syndrome and recurrent infections. J Clin Rheumatol. 2017;23:70–71.
  • Wang CR, Chiu YC, Chen YC. Successful treatment of refractory neutropenia in Felty’s syndrome with rituximab. Scand J Rheumatol. 2018;47:340–341.
  • Amezcua-Guerra LM, Springall R, Marquez-Velasco R, et al. Presence of antibodies against cyclic citrullinated peptides in patients with ‘rhupus’: a cross-sectional study. Arthritis Res Ther. 2006;8:R144.
  • Li J, Wu H, Huang X, et al. Clinical analysis of 56 patients with rhupus syndrome: manifestations and comparisons with systemic lupus erythematosus: a retrospective case-control study. Medicine (Baltimore). 2014;93:e49.
  • Danion F, Sparsa L, Arnaud L, et al. Long-term efficacy and safety of antitumour necrosis factor alpha treatment in rhupus: an open-label study of 15 patients. RMD Open. 2017;3(2):e000555.
  • Shovman O, Tamar S, Amital H, et al. Diverse patterns of anti-TNF-α-induced lupus: case series and review of the literature. Clin Rheumatol. 2018;37:563–568.
  • Andrade-Ortega L, Irazoque-Palazuelos F, Muñóz-López S, et al. Efficacy and tolerability of rituximab in patients with rhupus. Reumatol Clin. 2013;9:201–205.
  • Piga M, Gabba A, Cauli A, et al. Rituximab treatment for ‘rhupus syndrome’: clinical and power-Doppler ultrasonographic monitoring of response. A longitudinal pilot study. Lupus. 2013;22:624–628.
  • Araújo F, Silva I, Sepriano A, et al. Off-label use of rituximab in systemic rheumatic diseases: case series and review. Acta Reumatol Port. 2013;38:290–294.
  • Ikeda K, Sanayama Y, Makita S, et al. Efficacy of abatacept for arthritis in patients with an overlap syndrome between rheumatoid arthritis and systemic lupus erythematosus. Clin Dev Immunol. 2013;2013:697525.
  • Brito-Zerón P, Baldini C, Bootsma H, et al. Sjögren syndrome. Nat Rev Dis Primers. 2016;2:16047.
  • Sankar V, Brennan MT, Kok MR, et al. Etanercept in Sjögren’s syndrome: a twelve-week randomized, double-blind, placebo-controlled pilot clinical trial. Arthritis Rheum. 2004;50:2240–2245.
  • Seror R, Sordet C, Guillevin L, et al. Tolerance and efficacy of rituximab and changes in serum B cell biomarkers in patients with systemic complications of primary Sjögren’s syndrome. Ann Rheum Dis. 2007;66:351–357.
  • Dass S, Bowman SJ, Vital EM, et al. Reduction of fatigue in Sjögren syndrome with rituximab: results of a randomised, double-blind, placebo-controlled pilot study. Ann Rheum Dis. 2008;67:1541–1544.
  • Meijer JM, Meiners PM, Vissink A, et al. Effectiveness of rituximab treatment in primary Sjögren’s syndrome: a randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 2010;62:960–968.
  • Sada PR, Isenberg D, Ciurtin C. Biologic treatment in Sjögren’s syndrome. Rheumatology (Oxford). 2015;54:219–230.
  • Carson SE, Vivino FB, Parke A, et al. Treatment guidelines for rheumatologic manifestations of Sjögren’s syndrome: use of biologic agents, management of fatigue, and inflammatory musculoskeletal pain. Arthritis Care Res (Hoboken). 2017;69:517–527.
  • Andréu Sánchez JL, Fernández Castro M, Del Campo Fontecha PD, et al. SER recommendations on the use of biological drugs in primary Sjögren’s syndrome. Reumatol Clin. 2019;15:315–326.
  • Lam GK, Hummers LK, Woods A, et al. Efficacy and safety of etanercept in the treatment of scleroderma-associated joint disease. J Rheumatol. 2007;34:1636–1637.
  • Hamaguchi M, Kawahito Y, Ishino H, et al. A case report of tumor necrosis factor-alpha antibody-induced thrombocytopenia associated with emerging IgM anticardiolipin antibody in patients with scleroderma overlap/rheumatoid arthritis. Clin Rheumatol. 2007;26:988–990.
  • Christopher-Stine L, Wigley F. Tumor necrosis factor-alpha antagonists induce lupus-like syndrome in patients with scleroderma overlap/mixed connective tissue disease. J Rheumatol. 2003;30:2725–2727.
  • Bosello S, De Luca G, Tolusso B, et al. B cells in systemic sclerosis: a possible target for therapy. Autoimmun Rev. 2011;10:624–630.
  • Atzeni F, Sarzi-Puttini P, Botsios C, et al. Long-term anti-TNF therapy and the risk of serious infections in a cohort of patients with rheumatoid arthritis: comparison of adalimumab, etanercept and infliximab in the GISEA registry. Autoimmun Rev. 2012;12:225–229.
  • Schiff M, Weinblatt ME, Valente R, et al. Head-to-head comparison of subcutaneous abatacept versus adalimumab for rheumatoid arthritis: two-year efficacy and safety findings from AMPLE trial. Ann Rheum Dis. 2014;73:86–94.
  • Weinblatt ME, Moreland LW, Westhovens R, et al. Safety of abatacept administered intravenously in treatment of rheumatoid arthritis: integrated analyses of up to 8 years of treatment from the abatacept clinical trial program. J Rheumatol. 2013;40:787–797.
  • Alten R, Kaine J, Keystone E, et al. Long-term safety of subcutaneous abatacept in rheumatoid arthritis: integrated analysis of clinical trial data representing more than four years of treatment. Arthritis Rheum. 2014;66:1987–1997.
  • Yun H, Xie F, Delzell E, et al. Risk of hospitalised infection in rheumatoid arthritis patients receiving biologics following a previous infection while on treatment with anti-TNF therapy. Ann Rheum Dis. 2015;74:1065–1071.
  • Zhao S, Mysler E, Moots RJ. Etanercept for the treatment of rheumatoid arthritis. Immunotherapy. 2018;10:433–445.
  • 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;75:795–810.
  • Porter C, Armstrong-Fisher S, Kopotsha T, et al. Certolizumab pegol does not bind the neonatal Fc receptor (FcRn): consequences for FcRn-mediated in vitro transcytosis and ex vivo human placental transfer. J Reprod Immunol. 2016;116:7–12.
  • 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;77:228–233.
  • Clowse ME, Wolf DC, Förger F, et al. Pregnancy outcomes in subjects exposed to certolizumab pegol. J Rheumatol. 2015;42:2270–2278.
  • Clowse MEB, Scheuerle AE, Chambers C, et al. Pregnancy outcomes after exposure to certolizumab pegol: updated results from a pharmacovigilance safety database. Arthritis Rheumatol. 2018;70:1399–1407.
  • Clowse ME, Förger F, Hwang C, et al. Minimal to no transfer of certolizumab pegol into breast milk: results from CRADLE, a prospective, postmarketing, multicentre, pharmacokinetic study. Ann Rheum Dis. 2017;76:1890–1896.
  • Prieto-Peña D, Calderón-Goercke M, Adán A, et al. Efficacy and safety of certolizumab pegol in pregnant women with uveitis. Recommendations on the management with immunosuppressive and biologic therapies in uveitis during pregnancy. Clin Exp Rheumatol. 2020. online ahead of print.
  • Hyrich KL, Verstappen SM. Biologic therapies and pregnancy: the story so far. Rheumatology (Oxford). 2014;53:1377–1385.
  • Cantini F, Nannini C, Niccoli L, et al. Guidance for the management of patients with latent tuberculosis infection requiring biologic therapy in rheumatology and dermatology clinical practice. Autoimmun Rev. 2015;14:503–509.
  • Terrault NA, Lok ASF, McMahon BJ, et al. Update on prevention, diagnosis, and treatment of chronic hepatitis B: AASLD 2018 hepatitis B guidance. Hepatology. 2018;67:1560–1599.
  • Ghany MG, Morgan TR. AASLD-IDSA hepatitis c guidance panel. Hepatitis C guidance 2019 update: American Association for the study of liver diseases-infectious diseases society of America recommendations for testing, managing, and treating hepatitis C virus infection. Hepatology. 2020;71:686–721.
  • Zerbini CAF, Clark P, Mendez-Sanchez L, et al. Biologic therapies and bone loss in rheumatoid arthritis. Osteoporos Int. 2017;28:429–446.
  • Di Munno O, Ferro F. The effect of biologic agents on bone homeostasis in chronic inflammatory rheumatic diseases. Clin Exp Rheumatol. 2019;37:502–507.
  • Orsolini G, Fassio A, Rossini M, et al. Effects of biological and targeted synthetic DMARDs on bone loss in rheumatoid arthritis. Pharmacol Res. 2019;147:104354.
  • Mercer LK, Lunt M, Low AL, et al. Risk of solid cancer in patients exposed to anti-tumour necrosis factor therapy: results from the British Society for Rheumatology Biologics Register for Rheumatoid Arthritis. Ann Rheum Dis. 2015;74:1087–1093.
  • Wadström H, Frisell T, Askling J. Anti-Rheumatic Therapy in Sweden (ARTIS) Study Group. Malignant neoplasms in patients with rheumatoid arthritis treated with Tumor Necrosis Factor inhibitors, Tocilizumab, Abatacept, or Rituximab in clinical practice: A nationwide cohort study from Sweden. JAMA Intern Med. 2017;177:1605–1612.
  • Regierer AC, Strangfeld A. Rheumatoid arthritis treatment in patients with a history of cancer. Curr Opin Rheumatol. 2018;30:288–294.
  • Xie W, Xiao S, Huang Y, et al. A meta-analysis of biologic therapies on risk of new or recurrent cancer in patients with rheumatoid arthritis and a prior malignancy. Rheumatology (Oxford). 2020;59:930–939.
  • Available from: https://www.rheumatology.org/Portals/0/Files/Rheumatoid-Arthritis-Guideline-Project-Plan.pdf  (Accessed date: 20 October 2020).

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.