ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Interleukin 6 inhibitors: Biology, principles of use, and adverse effects

Interleukin 6 inhibitors: Biology, principles of use, and adverse effects
Literature review current through: Jan 2024.
This topic last updated: May 02, 2022.

INTRODUCTION — Interleukin (IL) 6 is a potent proinflammatory mediator that is important in immune defense and in immune-mediated disease. Four pharmacologic inhibitors (sometimes termed blockers) of IL-6 are commercially available for clinical use: tocilizumab, sarilumab, and satralizumab, which are all monoclonal antibodies directed against the IL-6 receptor (IL-6R), and siltuximab, a monoclonal antibody specific for IL-6.

General considerations regarding the use of these agents are presented here. The use of these agents for the treatment of specific inflammatory and autoimmune conditions is described in detail separately:

(See "Treatment of rheumatoid arthritis in adults resistant to initial biologic DMARD therapy", section on 'IL-6 inhibitor therapies' and "Treatment of rheumatoid arthritis in adults resistant to initial conventional synthetic (nonbiologic) DMARD therapy", section on 'Methotrexate plus IL-6 inhibitor/IL-6 inhibitor monotherapy'.)

(See "Polyarticular juvenile idiopathic arthritis: Treatment", section on 'Tocilizumab'.)

(See "Systemic juvenile idiopathic arthritis: Treatment", section on 'Interleukin 6 inhibitors'.)

(See "Treatment of adult-onset Still's disease", section on 'Interleukin 6 inhibition'.)

(See "Treatment of giant cell arteritis", section on 'Tocilizumab'.)

(See "HHV-8-negative/idiopathic multicentric Castleman disease", section on 'IL-6 inhibitors'.)

(See "Neuromyelitis optica spectrum disorder (NMOSD): Treatment and prognosis", section on 'Attack prevention'.)

INTERLEUKIN 6 BIOLOGY AND FUNCTION — Interleukin (IL) 6 is a proinflammatory cytokine, originally described as B cell differentiation factor, that mediates pleiotropic functions in immunologic responses during host infection, inflammatory disease, hematopoiesis, and oncogenesis. It can also exhibit antiinflammatory effects [1-3].

The IL-6 receptor (IL-6R) comprises two functional chains, IL-6R alpha (IL-6Ra) and a 130 kD non-ligand-binding chain capable of signal-transduction (glycoprotein 130 [gp130]), which mediate gene activation and a wide range of biologic activities (figure 1) [4,5]. IL-6 binds to both soluble and membrane-bound receptors, using three different but related signaling pathways [2,6,7]. It can signal by binding membrane-bound IL-6Ra/gp130 complexes on target cells (termed classic cis signaling) and can form soluble complexes with non-membrane bound IL-6Ra. Membrane expression of gp130 in the absence of IL-6Ra is widespread and confers upon the expressing cell the capacity to respond to circulating IL-6/IL-6Ra complexes (termed soluble trans-signaling). Trans-presentation of IL-6 by dendritic cell (DC)-bound IL-6Ra to gp130-expressing T cells can also induce a signal (termed trans presentation) [7,8].

IL-6Ra is expressed on a limited number of cell types, including hematopoietic cells and hepatocytes. By contrast, gp130 is expressed ubiquitously on most cells. The classic pathway is thought to induce the acute phase response, protect against infection, and stimulate metabolism and regeneration in liver and intestinal epithelium, while the soluble trans pathway is thought to be more important in stimulating inflammation.

IL-6 regulates the immune response, specifically the proliferation and differentiation of T cells and the terminal differentiation of B cells. IL-6 also activates macrophages and osteoclasts and is a pivotal mediator of the hepatic acute phase response. In concert with tumor necrosis factor (TNF) alpha and IL-1, IL-6 promotes vascular endothelial growth factor (VEGF) and metalloproteinase production [5]. IL-6, together with transforming growth factor (TGF) beta, also has a role in the development of regulatory T cells. IL-6, like IL-1, is important for both systemic and local inflammation that is often associated with symptoms like fever, fatigue, and anorexia, with changes in acute phase proteins in the plasma (eg, C-reactive protein and fibrinogen).

IL-6 has been implicated in the pathogenesis of autoimmune diseases. As an example, in rheumatoid arthritis (RA), IL-6 is expressed in RA synovial tissues and plays an important role in upregulation of endothelial adhesion molecule expression, in osteoclast maturation, and in promoting bone erosion. IL-6 also mediates systemic features of disease, including fatigue, cognitive dysfunction, and metabolic shift.

PRETREATMENT TESTING — We perform the following testing and evaluation before initiating treatment with an interleukin (IL) 6 inhibitor:

Patients are at risk for cytopenias, dyslipidemias, and abnormalities in liver function (see 'Adverse effects' below). Baseline laboratory studies typically include a complete blood count with differential and platelet count; liver function tests, including alanine aminotransferase (ALT) and aspartate aminotransferase (AST), bilirubin, and alkaline phosphatase; and a lipid panel, including total cholesterol, low-density lipoprotein, high-density lipoprotein, and triglycerides. Additional testing may be required for assessment of disease activity, which varies depending upon the disorder being treated. Testing varies with region in some cases (eg, alkaline phosphatase is not typically performed in Germany for these patients).

Females of reproductive age should be screened for pregnancy and counseled appropriately with respect to reproductive risk. Additional information regarding risk during pregnancy and breastfeeding is provided separately, including in the respective drug information topics for each agent. (See "Safety of rheumatic disease medication use during pregnancy and lactation", section on 'Tocilizumab'.)

Screening for latent tuberculosis, hepatitis B (including testing for hepatitis B virus [HBV] surface antigen and HBV core antibody), and hepatitis C virus is generally performed prior to initiating treatment. (See 'Infection' below and "Tuberculosis infection (latent tuberculosis) in adults: Approach to diagnosis (screening)" and "Tuberculosis infection (latent tuberculosis) in children" and "Hepatitis B virus: Screening and diagnosis in adults" and "Screening and diagnosis of chronic hepatitis C virus infection".)

In patients with rheumatoid arthritis (RA), giant cell arteritis (GCA), and systemic sclerosis with interstitial lung disease (SSc with ILD), IL-6 inhibitors, including tocilizumab and sarilumab, are usually avoided in those with absolute neutrophil counts <2000/mm3, platelet counts <100,000/mm3, or ALT or AST levels >1.5 times the upper limit of normal. Patients with cytopenias or abnormal liver testing who do receive these drugs should be closely monitored. (See "Treatment of rheumatoid arthritis in adults resistant to initial biologic DMARD therapy", section on 'IL-6 inhibitor therapies'.)

Treatment with IL-6 inhibitors is generally avoided in patients with prior diverticulitis, active hepatic disease or hepatic impairment, or in patients with RA or GCA whose baseline ALT or AST is >1.5 times the upper limit of normal. In patients in whom there is an urgent indication for use, such as resistant GCA, consultation with a gastroenterologist or a hepatologist is advised.

Patients with severe or life-threatening cytokine release syndrome (CRS) may exhibit cytopenias or elevated liver transaminases due to the underlying disease state and/or its treatment. Thus, relative risks and benefits should be evaluated on an individual basis in such patients. (See "Cytokine release syndrome (CRS)", section on 'Laboratory'.)

STRUCTURE AND MECHANISM OF ACTION OF SPECIFIC AGENTS

TocilizumabTocilizumab is a humanized immunoglobulin G subclass 1 (IgG1) monoclonal antibody to the interleukin (IL) 6 receptor (IL-6R). The antibody is made by grafting the complementarity determining regions of a mouse anti-human IL-6R monoclonal antibody onto human IgG1. Tocilizumab binds to both the membrane-bound and soluble forms of human IL-6R, thereby inhibiting the binding of the native cytokine to its receptor and interfering with the cytokine's effects.

SarilumabSarilumab is a fully human recombinant IgG1 monoclonal antibody that is directed against the membrane-bound and soluble forms of the IL-6R. As with tocilizumab, sarilumab inhibits the binding of the native cytokine to its receptor and interferes with the cytokine's effects.

SiltuximabSiltuximab is a chimeric anti-IL-6 monoclonal antibody that directly inhibits IL-6.

SatralizumabSatralizumab is a humanized monoclonal antibody that binds IL-6Rs, thereby suppressing inflammation mediated by IL-6 signaling pathways.

Investigational agents – Several agents that also inhibit IL-6 have been investigated for various disorders but are not available for clinical use. These include the anti-IL-6 antibodies, clazakizumab [9-11], and olokizumab [11-16].

ADVERSE EFFECTS

Range of adverse effects — The most commonly reported adverse events associated with interleukin (IL) 6 receptors (IL-6Rs)/IL-6 inhibitors are nasopharyngitis, headache, upper respiratory tract infection, gastritis, rash, arthralgia, extremity pain, fatigue, and nausea. Infections are the most frequent serious adverse events reported. Gastrointestinal perforation can occur in adults. Laboratory abnormalities include neutropenia, thrombocytopenia, dyslipidemia, and elevated liver enzymes. A significant increase in the rates of malignancy, autoimmunity, tuberculosis reactivation, or hepatitis have not been reported [17]. The risk of tuberculosis reactivation and of primary infection appears to be very low [18], although some trials excluded patients with latent tuberculosis.

Injection site reactions have been reported in approximately 10 percent of adults receiving tocilizumab by subcutaneous injection [19]; they may be more common in children. With intravenous administration, infusion reactions can occur, but anaphylaxis is rare [19,20]. Antidrug antibodies against tocilizumab occur in 0 to 16 percent of patients with rheumatoid arthritis (RA) and 1 to 8 percent of patients with juvenile idiopathic arthritis [21,22]. Loss of efficacy was not observed in the analysis of multiple trials involving nearly 9000 patients [22].

The safety of tocilizumab has been characterized in multiple randomized trials, systematic reviews, and meta-analyses, particularly for treatment of RA [17,23-29]. The adverse effects of other IL-6 inhibitors are thought to be similar, although there is less experience with sarilumab, as well as siltuximab, satralizumab, sirukumab [30-33], and clazakizumab.

Infection — In clinical trials and an integrated safety analysis of patients receiving an IL-6R inhibitor in combination with a conventional disease-modifying antirheumatic drug (DMARD) or as monotherapy, the risk of infections was found to be increased compared with placebo but in line with other biologic DMARDs [6,24,34-37].

An increased rate of infections, including mycobacterial and other opportunistic infections, is reported in patients treated with IL-6R/IL-6 inhibitors [24,26]. Most patients recover with appropriate treatment. The risk of serious infection is greater in patients on concomitant immunosuppressive therapy.

The reported serious infection rate in patients with RA treated with tocilizumab ranges from 3.7 to 5.7 per 100 patient-years [25,27,28]. Most infections are bacterial in nature, including pneumonia, bronchitis, cellulitis, and sepsis, although atypical mycobacterial infections, Pneumocystis jirovecii pneumonia, and herpes zoster infections occur as well. Risk factors include advanced age, long disease duration, concomitant therapy (such as glucocorticoids and methotrexate), and respiratory comorbidities. Higher rates of infections, including sinopulmonary infections, septic arthritis, varicella, and gastroenteritis, were also reported in a trial of tocilizumab in children with systemic juvenile idiopathic arthritis [38].

A 2011 meta-analysis of safety data in patients with RA on tocilizumab found that opportunistic infections developed at a rate of 0.23 episodes per 100 patient-years [26]. Such infections included active tuberculosis, nontuberculous mycobacteria, invasive candidiasis, P. jirovecii pneumonia, and cryptococcosis, especially with higher doses of tocilizumab. Another 2011 meta-analysis of randomized trials in patients with RA found that the rate of infection was increased in patients receiving tocilizumab plus methotrexate compared with placebo plus methotrexate (odds ratio [OR] 1.3, 95% CI 1.1-1.6) [17].

In patients with coronavirus disease 2019 (COVID-19), several observational studies found an association between treatment with tocilizumab and increased rates of fungal infections, particularly Candida and Aspergillus, and bacterial infections in patients with severe disease [39-41]. However, many of the patients in these studies were critically ill, and they also received glucocorticoids, sometimes in high doses, as well as other immunosuppressive agents. In addition, a meta-analysis of 10 observational studies of patients with COVID-19 did not show a clear association between tocilizumab and increased secondary infections [42]. Furthermore, this finding was not confirmed in several randomized trials, in which fewer serious infections were seen in patients hospitalized with COVID-19 who were receiving tocilizumab compared with placebo [43-45]. Despite lack of a clearcut signal for increased risk of fungal and bacterial infections in patients with COVID-19 treated with tocilizumab, clinicians should still monitor these patients closely for serious infections, particularly those with severe disease who have received other immunosuppressive agents.

Few serious infections have been reported in patients treated with siltuximab [46].

Hematologic — Neutropenia and thrombocytopenia have been reported and may require dose adjustment or drug discontinuation [29,38] according to recommendations in the summary of product characteristics (SPC) issued by the European Medicines Agency (EMA). Leukopenia, lymphopenia, and polycythemia have also been reported [46]. Whether the neutropenia is clinically relevant or is due to a margination of neutrophils to the vessel walls is still not clear.

Gastrointestinal

Dyslipidemia – IL-6 inhibitors may cause dyslipidemia [23,38,46]. In a systematic review, patients receiving tocilizumab were more likely to experience an elevation in the ratios of low-density lipoprotein to high-density lipoprotein cholesterol (20 versus 12 percent, relative risk [RR] 1.7, 95% CI 1.2-2.2) and of total to high-density lipoprotein cholesterol (12 versus 7 percent, RR 1.7, 95% CI 1.2-2.6) [23]. Dyslipidemia should be managed according to available guidelines. (See "Low-density lipoprotein cholesterol-lowering therapy in the primary prevention of cardiovascular disease" and "Management of low density lipoprotein cholesterol (LDL-C) in the secondary prevention of cardiovascular disease".)

Elevated liver enzymes – Significant liver enzyme (aminotransferase) elevations have been reported [38] and may require dose adjustment or drug discontinuation. Serious liver disease is rare.

Gastrointestinal symptoms and gastrointestinal tract perforation – Gastrointestinal symptoms are more common in patients on IL-6 inhibitors and are usually mild (eg, nausea, abdominal pain) [24,46]. However, gastrointestinal tract perforations have been reported, especially in older patients, particularly those on glucocorticoids, and in patients with a history of diverticulitis, which is a contraindication to the use of tocilizumab [29,47]. Patients should be instructed that these disorders may occur and that clinical manifestations may be blunted without a significant elevation of inflammatory laboratory parameters such as C-reactive protein. Some experts provide patients a note to inform clinicians in emergency departments about IL-6 blocking treatment and its potential effect on the clinical presentation in such instances.

EFFECTS ON ACUTE PHASE REACTANTS — Interleukin (IL) 6 inhibitors are antiinflammatory and suppress fever and acute phase reactants, such as C-reactive protein and ferritin. Masking of the signs and symptoms of acute inflammation can delay the diagnosis of infection, make it more difficult to diagnosis macrophage activation syndrome (MAS) in patients with systemic juvenile idiopathic arthritis, and can lead to overestimation of the clinical response to IL-6 inhibition if the composite disease activity score used gives relatively greater weight to levels of acute phase reactants. (See "HHV-8-negative/idiopathic multicentric Castleman disease", section on 'IL-6 inhibitors' and "Assessment of rheumatoid arthritis disease activity and physical function" and "Systemic juvenile idiopathic arthritis: Treatment", section on 'Biologic disease-modifying antirheumatic drug (DMARD) therapy' and "Systemic juvenile idiopathic arthritis: Course, prognosis, and complications", section on 'Diagnosis'.)

MONITORING AND RESTRICTIONS — Interleukin (IL) 6 inhibitors should not be given in combination with other biologic agents. Active infection should be excluded prior to every dose, and treatment should be delayed for patients with active infection until resolved. In addition, live or live-attenuated vaccines are not recommended during treatment. Patients should also be monitored for new onset of gastrointestinal symptoms that may suggest diverticulitis or gastrointestinal perforation and for signs and symptoms of central nervous system demyelinating disorders [6]. (See "General principles and overview of management of rheumatoid arthritis in adults", section on 'Drug monitoring and prevention of drug toxicity' and "Immunizations in autoimmune inflammatory rheumatic disease in adults" and "Neuromyelitis optica spectrum disorder (NMOSD): Treatment and prognosis", section on 'Attack prevention'.)

Recommended laboratory monitoring in patients on IL-6 inhibitors:

Rheumatoid arthritis (RA) in adults – Testing typically includes complete blood count with differential (neutrophils) and liver function tests (alanine aminotransferase [ALT], aspartate aminotransferase [AST], alkaline phosphatase, and bilirubin) every month until stable, then every three months. Testing varies in some regions (eg, alkaline phosphate is typically not performed in Germany). Lipids should be obtained every six months. (See "General principles and overview of management of rheumatoid arthritis in adults", section on 'Drug monitoring and prevention of drug toxicity'.)

Giant cell arteritis (GCA) – GCA is monitored in a similar fashion to RA.

Juvenile idiopathic arthritis (polyarticular and systemic) – Testing should include neutrophil and platelet counts, liver enzyme (AST/ALT) levels, and lipid levels at least monthly until stable and then every three months (six months for lipid levels). (See "Systemic juvenile idiopathic arthritis: Treatment", section on 'In patients on tocilizumab'.)

ADDITIONAL DISTINCTIONS BETWEEN AGENTS

Route of administrationTocilizumab can be administered subcutaneously or intravenously, siltuximab is only administered intravenously, and satralizumab and sarilumab are only administered subcutaneously. For additional information, refer to the drug interactions program included within UpToDate. These agents also differ by dosing intervals. As an example, for rheumatoid arthritis (RA), tocilizumab is given weekly (European Medicines Agency [EMA]) or every two weeks subcutaneously or every four weeks intravenously, whereas sarilumab is given every two weeks subcutaneously.

Mechanistic distinctionsTocilizumab, sarilumab, and satralizumab all bind to the interleukin (IL) 6 receptor (IL-6R), while siltuximab directly binds to IL-6 itself. It is not known whether there are particular advantages to targeting IL-6 itself rather than the receptor [2].

DISCONTINUING INTERLEUKIN 6 INHIBITION — In patients treated for rheumatoid arthritis (RA), discontinuation of tocilizumab, like most other biologic disease-modifying antirheumatic drugs (DMARDs), leads to a subsequent flare in most patients, even in those with low disease activity or remission [48-50]. Resumption of therapy improves disease control in the majority of patients.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Rheumatoid arthritis" and "Society guideline links: Juvenile idiopathic arthritis" and "Society guideline links: Side effects of anti-inflammatory and anti-rheumatic drugs".)

SUMMARY

Mechanisms of action – Interleukin (IL) 6 is a proinflammatory cytokine that mediates pleiotropic functions in immunologic responses during host infection, inflammatory disease, hematopoiesis, and oncogenesis. It can also exhibit antiinflammatory effects and has been implicated in autoimmune diseases. IL-6 binds to both soluble and membrane-bound forms of its receptor. (See 'Interleukin 6 biology and function' above.)

Pretreatment testing – Baseline laboratory studies should include a complete blood count, differential, and platelet count; liver function tests; and a lipid panel. Patients should also be screened for latent tuberculosis and hepatitis B and C. Females of reproductive age should be screened for pregnancy. (See 'Pretreatment testing' above.)

Types of agents available – Several agents are available for pharmacologic inhibition of IL-6, including the anti-IL-6 receptor (IL-6R) monoclonal antibodies tocilizumab, sarilumab, and satralizumab and the anti-IL-6 monoclonal antibody siltuximab. (See 'Structure and mechanism of action of specific agents' above.)

Adverse effects – Pharmacologic inhibition of IL-6 is generally well tolerated. The most commonly reported adverse events associated with IL-6 inhibitors are nasopharyngitis, headache, upper respiratory tract infection, gastritis, rash, arthralgia, extremity pain, fatigue, and nausea. Infections are the most frequent serious adverse events reported. Gastrointestinal perforation can occur in adults. Injection site and infusion reactions may occur. Laboratory abnormalities include neutropenia, thrombocytopenia, dyslipidemia, and elevated liver enzymes. (See 'Adverse effects' above.)

Effects on acute phase reactants – IL-6 inhibitors suppress fever and acute phase reactants, such as C-reactive protein and ferritin. (See 'Effects on acute phase reactants' above.)

Precautions – IL-6 inhibitors should not be given in combination with other biologic agents, and active infection should be excluded prior to every dose. Live or live-attenuated vaccines are not recommended during treatment. (See 'Adverse effects' above.)

Monitoring – Patients should be monitored for signs or symptoms of gastrointestinal or central nervous system complications. Laboratory monitoring includes blood counts, liver function tests, and lipid levels. (See 'Adverse effects' above.)

  1. Akira S, Taga T, Kishimoto T. Interleukin-6 in biology and medicine. Adv Immunol 1993; 54:1.
  2. Avci AB, Feist E, Burmester GR. Targeting IL-6 or IL-6 Receptor in Rheumatoid Arthritis: What's the Difference? BioDrugs 2018; 32:531.
  3. Choy EH, De Benedetti F, Takeuchi T, et al. Translating IL-6 biology into effective treatments. Nat Rev Rheumatol 2020; 16:335.
  4. Hirano T. Interleukin 6 and its receptor: ten years later. Int Rev Immunol 1998; 16:249.
  5. Okuda Y. Review of tocilizumab in the treatment of rheumatoid arthritis. Biologics 2008; 2:75.
  6. Favalli EG. Understanding the Role of Interleukin-6 (IL-6) in the Joint and Beyond: A Comprehensive Review of IL-6 Inhibition for the Management of Rheumatoid Arthritis. Rheumatol Ther 2020; 7:473.
  7. Heink S, Yogev N, Garbers C, et al. Trans-presentation of IL-6 by dendritic cells is required for the priming of pathogenic TH17 cells. Nat Immunol 2017; 18:74.
  8. Heink S, Yogev N, Garbers C, et al. Corrigendum: Trans-presentation of IL-6 by dendritic cells is required for the priming of pathogenic TH17 cells. Nat Immunol 2017; 18:474.
  9. Weinblatt ME, Mease P, Mysler E, et al. The efficacy and safety of subcutaneous clazakizumab in patients with moderate-to-severe rheumatoid arthritis and an inadequate response to methotrexate: results from a multinational, phase IIb, randomized, double-blind, placebo/active-controlled, dose-ranging study. Arthritis Rheumatol 2015; 67:2591.
  10. Mease PJ, Gottlieb AB, Berman A, et al. The Efficacy and Safety of Clazakizumab, an Anti-Interleukin-6 Monoclonal Antibody, in a Phase IIb Study of Adults With Active Psoriatic Arthritis. Arthritis Rheumatol 2016; 68:2163.
  11. Karkhur S, Hasanreisoglu M, Vigil E, et al. Interleukin-6 inhibition in the management of non-infectious uveitis and beyond. J Ophthalmic Inflamm Infect 2019; 9:17.
  12. Nasonov E, Fatenejad S, Feist E, et al. Olokizumab, a monoclonal antibody against interleukin 6, in combination with methotrexate in patients with rheumatoid arthritis inadequately controlled by methotrexate: efficacy and safety results of a randomised controlled phase III study. Ann Rheum Dis 2022; 81:469.
  13. Takeuchi T, Tanaka Y, Yamanaka H, et al. Efficacy and safety of olokizumab in Asian patients with moderate-to-severe rheumatoid arthritis, previously exposed to anti-TNF therapy: Results from a randomized phase II trial. Mod Rheumatol 2016; 26:15.
  14. Genovese MC, Fleischmann R, Furst D, et al. Efficacy and safety of olokizumab in patients with rheumatoid arthritis with an inadequate response to TNF inhibitor therapy: outcomes of a randomised Phase IIb study. Ann Rheum Dis 2014; 73:1607.
  15. Smolen JS, Feist E, Fatenejad S, et al. Olokizumab versus Placebo or Adalimumab in Rheumatoid Arthritis. N Engl J Med 2022; 387:715.
  16. Feist E, Fatenejad S, Grishin S, et al. Olokizumab, a monoclonal antibody against interleukin-6, in combination with methotrexate in patients with rheumatoid arthritis inadequately controlled by tumour necrosis factor inhibitor therapy: efficacy and safety results of a randomised controlled phase III study. Ann Rheum Dis 2022; 81:1661.
  17. Campbell L, Chen C, Bhagat SS, et al. Risk of adverse events including serious infections in rheumatoid arthritis patients treated with tocilizumab: a systematic literature review and meta-analysis of randomized controlled trials. Rheumatology (Oxford) 2011; 50:552.
  18. Rello J, Rodriguez R, Jubert P, Alvarez B. Severe community-acquired pneumonia in the elderly: epidemiology and prognosis. Study Group for Severe Community-Acquired Pneumonia. Clin Infect Dis 1996; 23:723.
  19. Burmester GR, Rubbert-Roth A, Cantagrel A, et al. A randomised, double-blind, parallel-group study of the safety and efficacy of subcutaneous tocilizumab versus intravenous tocilizumab in combination with traditional disease-modifying antirheumatic drugs in patients with moderate to severe rheumatoid arthritis (SUMMACTA study). Ann Rheum Dis 2014; 73:69.
  20. Park EH, Lee EY, Shin K, Kim HA. Tocilizumab-induced anaphylaxis in patients with adult-onset Still's disease and systemic juvenile idiopathic arthritis: a case-based review. Rheumatol Int 2020; 40:791.
  21. Strand V, Balsa A, Al-Saleh J, et al. Immunogenicity of Biologics in Chronic Inflammatory Diseases: A Systematic Review. BioDrugs 2017; 31:299.
  22. Burmester GR, Choy E, Kivitz A, et al. Low immunogenicity of tocilizumab in patients with rheumatoid arthritis. Ann Rheum Dis 2017; 76:1078.
  23. Singh JA, Beg S, Lopez-Olivo MA. Tocilizumab for rheumatoid arthritis: a Cochrane systematic review. J Rheumatol 2011; 38:10.
  24. Emery P, Keystone E, Tony HP, et al. IL-6 receptor inhibition with tocilizumab improves treatment outcomes in patients with rheumatoid arthritis refractory to anti-tumour necrosis factor biologicals: results from a 24-week multicentre randomised placebo-controlled trial. Ann Rheum Dis 2008; 67:1516.
  25. Nishimoto N, Miyasaka N, Yamamoto K, et al. Long-term safety and efficacy of tocilizumab, an anti-IL-6 receptor monoclonal antibody, in monotherapy, in patients with rheumatoid arthritis (the STREAM study): evidence of safety and efficacy in a 5-year extension study. Ann Rheum Dis 2009; 68:1580.
  26. Schiff MH, Kremer JM, Jahreis A, et al. Integrated safety in tocilizumab clinical trials. Arthritis Res Ther 2011; 13:R141.
  27. Yamamoto K, Goto H, Hirao K, et al. Longterm Safety of Tocilizumab: Results from 3 Years of Followup Postmarketing Surveillance of 5573 Patients with Rheumatoid Arthritis in Japan. J Rheumatol 2015; 42:1368.
  28. Curtis JR, Perez-Gutthann S, Suissa S, et al. Tocilizumab in rheumatoid arthritis: a case study of safety evaluations of a large postmarketing data set from multiple data sources. Semin Arthritis Rheum 2015; 44:381.
  29. Sepriano A, Kerschbaumer A, Smolen JS, et al. Safety of synthetic and biological DMARDs: a systematic literature review informing the 2019 update of the EULAR recommendations for the management of rheumatoid arthritis. Ann Rheum Dis 2020; 79:760.
  30. Takeuchi T, Thorne C, Karpouzas G, et al. Sirukumab for rheumatoid arthritis: the phase III SIRROUND-D study. Ann Rheum Dis 2017; 76:2001.
  31. Aletaha D, Bingham CO 3rd, 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 2017; 389:1206.
  32. Food and Drug Administration Center for Drug Evaluation and Research. Summary Minutes of the Arthritis Advisory Committee Meeting. August 2, 2017 https://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/ArthritisAdvisoryCommittee/UCM575678.pdf (Accessed on October 02, 2018).
  33. FDA Briefing Document. Arthritis Advisory Committee Meeting August 2, 2017 https://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/ArthritisAdvisoryCommittee/UCM569150.pdf (Accessed on October 02, 2018).
  34. 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.
  35. Fleischmann R, Genovese MC, Lin Y, et al. Long-term safety of sarilumab in rheumatoid arthritis: an integrated analysis with up to 7 years' follow-up. Rheumatology (Oxford) 2020; 59:292.
  36. 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.
  37. 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.
  38. De Benedetti F, Brunner HI, Ruperto N, et al. Randomized trial of tocilizumab in systemic juvenile idiopathic arthritis. N Engl J Med 2012; 367:2385.
  39. Górgolas Hernández-Mora M, Cabello Úbeda A, Prieto-Pérez L, et al. Compassionate use of tocilizumab in severe SARS-CoV2 pneumonia. Int J Infect Dis 2021; 102:303.
  40. Kimmig LM, Wu D, Gold M, et al. IL6 inhibition in critically ill COVID-19 patients is associated with increased secondary infections. medRxiv 2020.
  41. Maeda T, Obata R, Rizk DO D, Kuno T. The association of interleukin-6 value, interleukin inhibitors, and outcomes of patients with COVID-19 in New York City. J Med Virol 2021; 93:463.
  42. Malgie J, Schoones JW, Pijls BG. Decreased Mortality in Coronavirus Disease 2019 Patients Treated With Tocilizumab: A Rapid Systematic Review and Meta-analysis of Observational Studies. Clin Infect Dis 2021; 72:e742.
  43. Stone JH, Frigault MJ, Serling-Boyd NJ, et al. Efficacy of Tocilizumab in Patients Hospitalized with Covid-19. N Engl J Med 2020; 383:2333.
  44. Hermine O, Mariette X, Tharaux PL, et al. Effect of Tocilizumab vs Usual Care in Adults Hospitalized With COVID-19 and Moderate or Severe Pneumonia: A Randomized Clinical Trial. JAMA Intern Med 2021; 181:32.
  45. Salvarani C, Dolci G, Massari M, et al. Effect of Tocilizumab vs Standard Care on Clinical Worsening in Patients Hospitalized With COVID-19 Pneumonia: A Randomized Clinical Trial. JAMA Intern Med 2021; 181:24.
  46. van Rhee F, Casper C, Voorhees PM, et al. A phase 2, open-label, multicenter study of the long-term safety of siltuximab (an anti-interleukin-6 monoclonal antibody) in patients with multicentric Castleman disease. Oncotarget 2015; 6:30408.
  47. Strangfeld A, Richter A, Siegmund B, et al. Risk for lower intestinal perforations in patients with rheumatoid arthritis treated with tocilizumab in comparison to treatment with other biologic or conventional synthetic DMARDs. Ann Rheum Dis 2017; 76:504.
  48. Bouman CAM, Tweehuysen L, Haverkort D, et al. Abatacept and tocilizumab tapering in rheumatoid arthritis patients: results of SONATA-a retrospective, exploratory cohort study. Rheumatol Adv Pract 2018; 2:rky008.
  49. Huizinga TW, Conaghan PG, Martin-Mola E, et al. Clinical and radiographic outcomes at 2 years and the effect of tocilizumab discontinuation following sustained remission in the second and third year of the ACT-RAY study. Ann Rheum Dis 2015; 74:35.
  50. Aguilar-Lozano L, Castillo-Ortiz JD, Vargas-Serafin C, et al. Sustained clinical remission and rate of relapse after tocilizumab withdrawal in patients with rheumatoid arthritis. J Rheumatol 2013; 40:1069.
Topic 129345 Version 15.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟