Open access peer-reviewed chapter

Autoantibodies and Cytokines in Pathogenesis of Rheumatoid Arthritis

Written By

Mohd Wajid Ali Khan and Wahid Ali Khan

Submitted: 27 March 2018 Reviewed: 26 October 2018 Published: 22 November 2018

DOI: 10.5772/intechopen.82265

From the Edited Volume

Autoantibodies and Cytokines

Edited by Wahid Ali Khan

Chapter metrics overview

1,304 Chapter Downloads

View Full Metrics

Abstract

Rheumatoid arthritis (RA) is an autoimmune disorder in which increased autoantibody production and enhanced secretion of pro-inflammatory cytokines are the hallmark of the disease. A strictly controlled balance of antibody production and proinflammatory cytokines is the key to the healthy state. A slight tilt in this balance causes proinflammatory diseases. In RA there is an increased production of autoantibodies such as rheumatoid factor (RF) and anti-citrullinated protein antibody (ACPA), anti-cartilage type II antibodies, and etc., which have a prominent clinical significance. Furthermore, there is increased secretion of proinflammatory cytokines such as tumor necrosis factor-α (TNFα), interleukin-6 (IL-6), interleukin-1 (IL-1) which have an impact of great magnitude on the RA disease progression and severity. A better understanding of the mechanism of autoantibody production and secretion of cytokines together with crosstalk between immune cells and cytokines can provide us a better insight into the disease pathogenesis as well disease prognosis and management.

Keywords

  • rheumatoid arthritis
  • autoantibodies
  • cytokines
  • proinflammatory
  • pathogenesis

1. Introduction

Autoimmunity arises as a result of failure of immune self-tolerance. The condition may involve both T and B cells however it has been found that in most autoimmune diseases, T cells play a pivotal role in both dysregulation and autoimmune aggression, but autoantibodies are also widely produced by B cells. Such autoantibodies play a key pathogenic role in diseases such as autoimmune hemocytopenias, Grave’s disease, rheumatoid arthritis, type 1 diabetes and systemic lupus erythemathosus (SLE). Similarly, autoantibodies are also found in other diseases, where although they may play a minor pathogenic role, but can be used as valuable diagnostic markers [1, 2, 3, 4, 5, 6, 7].

Furthermore, in addition to T cells and autoantibodies, cytokines also play a pivotal role in development of the autoimmune response. Proinflammatory cytokines have significant involvement in autoimmune associated damage. This chapter aims to discuss the involvement of autoantibodies and cytokines in the pathogenesis of RA.

Immune system has the capacity to mount an immune response against virtually all foreign molecules as well as self. However, several mechanisms exist within the human system that prevent or subdue the response to self-antigens. The immune system has developed a series of checks and balances that enable it to distinguish dangerous signals from harmless ones and allow it to respond to foreign or non-self-antigens. When these mechanisms undergo a breakdown or are overridden, a response directed against self-antigen can occur, resulting in autoimmune reactions and/or autoimmune diseases [8].

RA is a complex chronic disease, primarily affects the lining of the synovial joints and can cause progressive disability, premature death, and socioeconomic burdens. The clinical manifestations of symmetrical joint involvement include arthralgia, swelling, redness, and even limiting the range of motion [9].

Autoantibodies have been associated with human pathologies for a long time, particularly with autoimmune diseases. RA is more frequent in females as compared to males [10]. Organ specific autoimmune diseases involve single or multiple autoantigens. In RA, presence of various autoantibodies such as RF and ACPA, and anti-cartilage type II antibodies in serum and synovial fluid have long been associated with RA severity [11].

The pathological hallmarks of synovitis in rheumatoid arthritis include the proliferation of resident synovial fibroblasts, new blood vessel formation and the recruitment of a wide range of leukocytes including B and T lymphocytes, monocytes/macrophages and mast cells; in turn this leads to synovial hypertrophy and the invasion of cartilage and bone by activated inflammatory tissue. Cytokines are fundamental orchestrators of the development and maintenance of this lesion [12]. RA disorder is a multifactorial disorder other than autoantibodies there are many other important factors involved such as proinflammatory cytokines such as TNF-α, IL-6 and IL-1 are key mediators of cell migration and inflammation in RA [13]. Cartilage degradation in RA occurs when TNF-α, IL-1 and IL-6 activate synoviocytes, resulting in the secretion of matrix metalloproteinases (MMPs), cathepsins and mast cell proteinases into the synovial fluid [14, 15]. Cytokines also activate chondrocytes, leading to the direct release of additional MMPs into the cartilage [14, 15].

Although the availability of advanced drugs and treatment regimes, however the complete remission of the disease is still not achieved. This chapter shed light on the complex network of the autoantibodies and proinflammatory cytokines as immune responses in the RA disease pathogenesis and the development of bio-therapeutics used in RA disorder.

Advertisement

2. Autoantibodies in RA

Several studies have been demonstrated that levels of disease-related biomarkers (such as RF and antibodies to citrullinated protein antigen, as well as secretory phospholipase A2, C-reactive protein (CRP), glycated HSA, and multiple cytokines/chemokines) may be elevated prior to the onset of symptomatic rheumatoid arthritis [16, 17, 18, 19, 20, 21, 22, 23, 24].

These findings suggest that there is a substantial “preclinical” period of RA, during which detectable immunologic and inflammatory changes are occurring that are related to disease development. These increased levels of RA related autoantibodies in preclinical RA may be highly specific for early RA detection [18, 19]. There is a great hope that these autoantibodies may be used to predict which currently asymptomatic individuals are at sufficiently high risk for future RA that they may be targeted with preventive therapies.

2.1. Rheumatoid factors

The RA is associated with systemic autoimmunity as evidenced by the presence of serum and synovial fluid autoantibodies. The first autoantibody to be described in RA was the rheumatoid factor (RF) by Waaler in 1940 [25], and it was later found to be directed to the Fc region of IgG. It is well characterized, although its exact origin still remains unclear. Typically, RF is of IgM isotype, but IgG and IgA may also occur. IgM RFs are the major RF species in RA and are detected in 60–80% of RA patients [26]. In the past, RF levels were determined by classical agglutination reactions; however, sensitivities and specificities depended on the type of test (e.g., latex fixation test, or Waaler-Rose test using sheep erythrocytes). RF levels were also determined by nephelometry [27]. RF has been observed in many other autoimmune diseases, such as, in systemic lupus erythematosus, mixed connective tissue disease and primary Sjogren syndrome, as well as in non-autoimmune conditions, such as in chronic infections and old age [26]. RF specificity to RA is increased at high titers (e.g., IgM RF > 50 IU/ml) and with IgA isotypes [26, 28, 29]. High titer RF and IgA isotypes are also associated with radiologic erosion, extra-articular manifestations and thus, poorer outcomes [26, 28, 30, 31]. The association between high titer RF status and a poor prognosis indicates that RF may have a role in the pathogenesis of RA. The functions of RFs under normal physiological conditions were observed as (i) enhancement of immune complex clearance by increasing its avidity and size, (ii) aiding B cells in uptake of immune complex through efficient antigen presentation to T cells, and (iii) facilitation of complement fixation by binding to IgG containing immune complexes [32, 33, 34]. RFs with high affinity and high-titer in synovial fluid of RA patients are considered to exert pathogenic functions and to enhance inflammation and antigen trapping in joints. However, no clear evidence yet suggests that RFs are involved in the initial events triggering the disease process of RA. In fact, it is understood that they may themselves be triggered by RA. Somatic mutations accumulates in RA and the presence of isotype switching indicate that RF production is T-cell driven, although T cells infiltrate RA synovium [35] and contain autoreactive clones [36], which were polyclonal and lack specificity for any particular autoantigen [36, 37]. T-cell clones reactive with autologous IgG were not detected in RA patients as yet. Additionally, the function of RF expressing B cells to take up immune complexes and present trapped antigens to T cells may allow these cells to bypass the need for specific T cell help and eventually lead to emergence of autoreactive T cells capable of triggering RF synthesis in the absence of an external antigen [38].

2.2. Anti-citrullinated proteins antibodies (ACPA) in RA

Some other names used to describe ACPA are anti-keratin, antiperinuclear factor antibodies, antifilaggrin antibodies, or anti-Sa [39]. ACPA have been associated with human pathology [40] as well as preclinical disease [16, 18, 41]. Latest ELISA assays, exhibited higher specificity (∼98%) and sensitivity between 40 and 76% (depending on disease stage) [42]. Recently proved that there is a potential association of ACPA with conditions like psoriatic arthritis [43], periodontitis [44], and osteoarthritis [45]. The key difference between ELISA assays was in the antigens used to detect ACPA. Thus, the diagnostic value of ACAP was established by demonstrating the significance of using appropriate citrullinated peptide [39, 46, 47]. Consequentially, a highly sensitive noncommercial ELISA, based on protein targets identified as reactive with ACPA in synovial tissue such as alpha and beta fibrinogen was therefore developed [48]. The positivity of ACPA for one or both to these two citrullinated peptides covered all reactivity in RA sera [49].

Citrullinated peptides are generated in response to a posttranslational modification mediated by peptidyl-arginine deiminase (PAD) enzymes. Multiple antibody isotypes including IgG, IgA, and IgM directed against these citrullinated peptides are detected in RA [50]. Citrullinated proteins are present in the synovial fluid of inflamed RA joints, exhibiting that ACPA could bind to these antigens in the joint and possibly increase local inflammation [51]. A protein that is commonly targeted by ACPA is Vimentin. In collagen-induced arthritis, mouse models passive transfer of ACPA cannot cause synovitis, although it can worsen preexistent synovitis [52]. Therefore, it is suggested that multiple events are necessary for the development of RA.

ACPA causes inflammation via binding to Fc receptors or complement activation. Autoantibodies are usually glycoproteins that means both Fc and Fab region of the antibody bind to the carbohydrate chains, which is essential for immune effector functions. Compared to IgG antibodies, the Fc region of ACPA has a lower level of galactosylation and sialylation against recall antigens [53]. The decreased sialylation of IgG in immune complexes can drive osteoclastogenesis, both in vitro and in vivo, through altered FcγR signaling. Moreover, it has been found that RA patients with low levels of ACPA-IgG Fc sialylation displayed lower bone volumes and trabecula numbers [54]. Thus, disease pathophysiology could be influenced by the specific Fc glycan signature of ACPA.

2.3. Autoantibodies against type II collagen

Type II collagen (CII) are abundantly present in joint cartilage [55]. Native CII protein consists of a triple-helix structure containing three identical α chains. The collagen fibrils contribute to cartilage integrity by resisting stretching forces caused by hydrophilic proteoglycan molecules in extracellular matrix of articular cartilage, [56]. The degradation of CII leads to the degeneration of cartilage and consequent loss of function in RA patients [57]. Denaturation of CII also causes separation of α chains and loss of antigenic sites (epitopes) present in the molecule which are altered due to the disruption of its three dimensional structure [58]. Autoantibodies to both, native and denatured CII, have been reported in RA [59, 60, 61, 62]. Varying levels of anti-CII antibodies were detected in the same patient at different times and also between patients, suggesting that these antibodies might be associated with specific events during arthritis development.

Increased anti-CII antibody levels may degrade CII molecules leading to acute inflammation which is mediated by anti-CII antibody containing surface-bound immune complexes (ICs) [60], which activates complement system activation and enhanced the production of proinflammatory cytokines (TNFα, IL-1β and IL-8) [63], which leads to the inflammation of the joints and hence cartilage damage. Antibodies have been detected against major CII epitopes at the site of inflammation, serum and synovial fluid samples from RA patients, supporting the concept of an increased local immune response to CII in the joints [61].

CII is found to be arthritogenic in animals and an injection of native CII in adjuvant induces collagen-induced arthritis (CIA) characterized by antibodies to CII and inflammatory polyarthritis [64]. Variability in expression of arthritis is linked to the expression of particular class II major histocompatibility (MHC) alleles [65] and also depends on an intact immune system. For example, B cell deficient animals [66] or complement deficient ones [67] are protected. Moreover, monoclonal antibodies (mAb) to CII derived from mice with CIA can induce collagen antibody-induced arthritis (CAIA) in naïve mice. CAIA is a condition characterized by inflammation, formation of pannus and erosions of bone similar to that observed in RA [68]. This model disease does not require the help pf T cells and has proven to be an informative model to better understand how antibodies lead to development of arthritis. Not only is arthritis not MHC-restricted, but it can be induced in most strains of mice and represents a model of the effector arm of CIA. However, it depends on the specificity of the antibodies used. A triplet of arginine-glycine-hydrophobic acids, is a common amino acid motif, shared by these arthritogenic mAb, which recognize epitopes on CII. These map to surface-exposed regions on the collagen fibrils that are accessible for antibody binding [68]. These epitopes are conserved, and are also recognized by antibodies from rats [69, 70, 71] and from humans with RA [70, 72]. Amino acid arginine molecules are present on the surface of the major epitopes on the collagen fibrils can also become citrullinated [73] and mAb reactive with these citrullinated epitopes may be arthritogenic themselves, or induce more severe arthritis when injected with subclinical doses of anti-CII [74]. Antibodies to major CII epitopes could be useful as markers for the biomonitoring of joint destruction in some patients.

Advertisement

3. Role of cytokines in RA

The network of cytokine in the RA disease is very complex system, with a numerous of cytokines showing pleiotropic actions and many different targets. This network can be divided in two groups, the pro-inflammatory and anti-inflammatory cytokines. Controlling the balance between these two groups is considered as an important therapeutic goal. This chapter provide an important role of TNF-α in the pathogenesis of RA, leading to the first clinical trials of a biological therapeutic in this disease. Other than TNF-α we address other cytokines such as IL-1, IL-6 and IL-23 that might play a role in the disease, together with selected cytokines that bind a receptor containing the common γ-chain (γc) [75].

3.1. TNF-α

TNF-α is a proinflammaotory cytokine and played a key role in RA with its potential to degrade cartilage [76] and bone [77] in vitro. It has been shown in an experiment that dissociated RA synovial mononuclear cell cultures that TNF-α as well as other proinflammatory cytokines (IL-1, IL-6, GM-CSF, and IL-8) [78, 79, 80, 81] were produced in a five-day culture [82, 83]. When the activity of TNF-α was blocked in these cultures, the spontaneous production of both IL-1 protein and IL1B mRNA was remarkably decreased and IL-1 bioactivity was neutralized [82]. This is the evidence that the secretion of all these cytokines is a network and controlled by hierarchy of their expressions.

Soluble TNF receptors are found in high concentrations in the synovial fluid and serum of patient with RA [84]. RA patients are found to have high levels of TNF-α in the synovial fluid. This plays an important role in inflammation and joint destruction, both of which are hallmarks of RA. Anti-TNF-α therapy induces a shift in the cytokine equilibrium producing more anti-inflammatory cytokines. Studies have demonstrated dramatic improvement in synovial inflammation in RA patients after treatment with neutralizing anti-TNF-α Abs or soluble TNF receptors. They also suggest decreased joint destruction after treatment with IL-1Ra [85].

In a first clinical trial of a TNF-α blocking agent for the treatment of 20 active RA patients were initiated. Infliximab (Remicade), a chimeric antibody specific for human TNF-α was used. Signs and symptoms of the RA disease were substantially reduced with the treatment with infliximab together with decreased levels of CRP in the serum [86]. Other multicentric placebo-controlled trials were also confirmed the therapeutic efficacy of infliximab when coadministrated with methotrexate. This led eventually to FDA approval of the drug for the treatment of RA [87, 88]. After two-year of clinical trial, it was observed that there was a retardation or arrest of both joint space narrowing and bone erosion due to infliximab and methotrexate therapy [89]. There were two other drugs etanercept (Enbrel) and adalimumab (Humira) which are functioned as TNF-α blockers were used in the treatment of RA.

TNF-α is now considered as controlling a wide variety of effector functions relevant to the pathogenesis of RA, including endothelial cell activation and chemokines production which causes accumulation of leukocytes [90]; osteoclast and chondrocyte activation, promoting articular destruction. These all are RA disease pathogenesis spectrum which explains the broad role of TNF-α blockade in patients. Further, improved therapies targeting TNF-α would be a potential therapeutics for the treatment of RA.

3.2. Interleukin-1

Each member of the IL-1 family binds with high affinity to specific receptors. Binding of IL-1α or IL-1β to type I IL-1 receptors (IL-1RI), can is enhanced by an accessory protein, IL-1R-AcP, leads to intracellular signal transduction and regulation of gene expression and hence cellular responses [91]. The extent of response of IL-1β in the rheumatoid joint depends on a few factors such as (1) IL-1β and IL-1Rα have similar affinity for IL-1RI on synoviocytes, chondrocytes and other cells and hence, the relative concentrations of IL-1β and IL-1Rα are important in determining the level of cell activation and biological responses, (2) a greater number of IL-1RII reduces the amount of IL-1β and IL-1Rα, that is available for binding to IL-1RI. Similarly, soluble IL-1 receptors found in synovial fluid and in the circulation also decrease the amount of these cytokines available to interact with IL-1RI. The response of IL-1β, as well as to other proinflammatory cytokines, is regulated by various anti-inflammatory and immunomodulatory cytokines, including IL-4, IL-10, IL-11, IL-13 and transforming growth factor-β [91, 92].

IL1 causes inflammatory cells to move into the joints and the synovium in RA patients. An unspecified antigenic trigger is thought to activate the production of IL1 in joints by macrophages (lymphocytes, monocytes and transformed fibroblasts) [54, 93]. These cells secrete proteases and proteoglycans as cellular signals, that may result in pannus formation, which accumulates in the joints. Destructive enzymes can enter and destroy cartilage and ultimately degrade and erode bone. Importantly, specifically blocking IL1 is a targeted, rational treatment against the destructive functions of IL1 in RA [54, 94].

Evidence from experimental studies in animal models of arthritis and from an x randomized controlled trials in patients with RA indicates that IL-1 plays an important role in RA pathogenesis, and that IL-1 inhibition with anakinra is effective in slowing further radiographic progression of the disease and hence models significantly reduces bone erosions and cartilage degradation [14]. It is important to elucidate that, whether slowing radiographic progression with these biological therapies will significantly improve long-term outcomes in RA.

3.3. Interleukin-6

IL-6 is an essential and multifunctional proinflammatory cytokine of the immune system and could be a key mediator for the development of many chronic inflammatory or autoimmune diseases including RA [95, 96].

It is well established that increased levels of autoantibodies are the characteristics of autoimmune RA and hence decreased levels of antibody producing B cells are might have a therapeutic efficacy demonstrates the impact of B-cell activity on synovial inflammation and joint damage. IL-6 stimulates B cells to differentiate into plasma cells to produce immunoglobulins [97]. IL-6 induces B-cell differentiation [98] and it has been established that B-cells induced antibody production [99].

Neutrophils can be directly activated by IL-6 through membrane-bound receptor IL-6R, which in turn help inflammation and joint destruction through the secretion of proteolytic enzymes and reactive oxygen intermediates [100]. An in vitro study with fibroblasts from patients with RA showed the role of IL-6 in actively encourage the recruitment of neutrophil by activated fibroblasts. Although untreated fibroblasts were able to recruit neutrophils, it was found that the recruitment was inhibited in the presence of anti-IL-6 antibody [101].

Osteoclasts are multinucleated cells formed by the fusion of mononuclear progenitors of the monocyte and macrophage family. These cells populate the synovial membranes of RA patients and are concentrated in bones [102, 103]. Macrophage derived osteoclastogenesis requires the presence of macrophage colony-stimulating factor. It results from the interaction of the RANK and the RANK ligand (RANKL) [102]. RANKL expression is regulated by pro-inflammatory cytokines such as TNF-α, IL-1, IL-6 and IL-17 [103].

The principal cause of bone erosion is the pannus, which is found at the interface with cartilage and bone. Angiogenesis is an important process in the formation and maintenance of pannus [104]. Vascular endothelial growth factor (VEGF), is an important angiogenic mediator which promotes the migration and proliferation of endothelial cells, as well as inducing vascular permeability and mediating inflammation [105]. Increased levels of VEGF correlate with disease activity in RA patients [106]. IL-6 in the presence of sIL-6R increased VEGF levels in cultured synovial fibroblasts from RA patients and anti-IL-6R antibody significantly reduced VEGF concentration [107].

Blocking antibodies were used with other agents as a combinational therapeutics for the treatment of RA. A humanized anti-IL-6R monoclonal antibody, tocilizumab (TCZ), used in a first clinical trial was conducted in patients with established RA [108]. A total of 45 patients were randomized to receive a single intravenous infusion of TCZ of 0.1, 1, 5, 10 mg/kg or placebo. Patients in the 5 and 10 mg/kg arms showed rapidly improvement in disease activity. CRP normalized after treatment in the 5 and 10 mg/kg treated patients confirming IL-6 as the dominant cytokine in generating the acute-phase response in patients with RA. Another, double-blind, placebo-controlled trial in 164 RA patients was conducted and demonstrates that the clinical response was maintained with repeated dosing of TCZ monotherapy [109]. A European study CHARISMA, examined the combinational effect of TCZ with methotrexate (MTX). In the study of 359 RA patients with partial response to MTX, it was found that TCZ was efficacious as monotherapy or in combination with MTX although the latter appeared to enhance the benefit of TCZ [110]. There were many other clinical trials and studies that ensures the use of TCZ alone or in combinational therapies such as MTX results in sustained improvement in physical function and reduced radiographic joint damage in RA [111, 112, 113, 114, 115, 116, 117].

Advertisement

4. Conclusion

RA is a complex disease that develops through a series of events often referred to as disease continuum. It is an autoimmune and inflammatory disease that can be further aggravated with the increased production and secretion of autoantibodies (RF ACPA, anti-cartilage type II antibodies) and the secretion of proinflammatory cytokines (TNF, IL-1, IL-6). With a better knowledge and understanding of the crosstalk between the molecules involved in RA disease pathogenesis, it would be easier to identify better markers for RA disease as well as design and administer specific and efficient therapeutics that can control RA pathogenesis and deliver long term and permanent remission of the disease.

References

  1. 1. Özen S, Berk Ö, Şimşek DG, Darcan S. Clinical course of Hashimoto’s thyroiditis and effects of levothyroxine therapy on the clinical course of the disease in children and adolescents. Journal of Clinical Research in Pediatric Endocrinology. 2011;3:192-197
  2. 2. McKee A, Peyerl F. TSI assay utilization: Impact on costs of Graves’hyperthyroidism diagnosis. The American Journal of Managed Care. 2012;18:e1-e14
  3. 3. Sblattero D, Berti I, Trevisiol C, Marzari R, Tommasini A, Bradbury A, et al. Human recombinant tissue transglutaminase ELISA: An innovative diagnostic assay for celiac disease. The American Journal of Gastroenterology. 2000;95:1253-1257
  4. 4. Sultan Alouffi S, Sherwani S, Al-Mogbel MS, Sherwani MKA, Khan MWA. Depression and smoking augment the production of circulating autoantibodies against glycated HSA in rheumatoid arthritis patients. International Archives of Allergy and Immunology. 2018;29:1-11
  5. 5. Khan MWA, Qadrie ZL, Khan WA. Antibodies against gluco-oxidatively modified human serum albumin detected in diabetes-associated complications. International Archives of Allergy and Immunology. 2010;153:207-214
  6. 6. Khan, Sherwani S, Khan WA, Moinuddin, Ali R. Characterization of hydroxyl radical modified GAD65: A potential autoantigen in type 1 diabetes. Autoimmunity. 2009;42(2):150-158
  7. 7. Khan MWA, Banga K, Mashal SN, Khan WA. Detection of autoantibodies against reactive oxygen species modified glutamic acid decarboxylase-65 in type 1 diabetes associated complications. BMC Immunology. 2011;12:19
  8. 8. Khan FH. Autoimmunity and Autoimmune Diseases. The Elements of Immunology. India: Pearson Education; 2009
  9. 9. Guo Q, Wang Y, Xu D, Nossent J, Pavlos NJ, Xu J. Rheumatoid arthritis: Pathological mechanisms and modern pharmacologic therapies. Bone Research. 2018;6:15
  10. 10. Silman AJ, Pearson JE. Epidemiology and genetics of rheumatoid arthritis. Arthritis Research. 2002;4(3):S265-S272
  11. 11. McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. The New England Journal of Medicine. 2011;365(23):2205-2219
  12. 12. Ridgley LA, Anderson AE, Pratt AG. What are the dominant cytokines in early rheumatoid arthritis? Current Opinion in Rheumatology. 2018;30(2):207-214
  13. 13. Smolen JS, Aletaha D, Koeller M, Weisman MH, Emery P. New therapies for treatment of rheumatoid arthritis. Lancet. 2007;370:1861-1874
  14. 14. Strand V, Kavanaugh AF. The role of interleukin-1 in bone resorption in rheumatoid arthritis. Rheumatology. 2004;43(Suppl. 3):iii10-iii16
  15. 15. Johnson LL, Dyer R, Hupe DJ. Matrix metalloproteinases. Current Opinion in Chemical Biology. 1998;2:466-471
  16. 16. Kurki P, Aho K, Palosuo T, Heliovaara M. Immunopathology of rheumatoid arthritis: Antikeratin antibodies precede the clinical disease. Arthritis and Rheumatism. 1992;35:914-917
  17. 17. Aho K, Palosuo T, Heliovaara M, Knekt P, Alha P, von Essen R. Antifilaggrin antibodies within “normal” range predict rheumatoid arthritis in a linear fashion. The Journal of Rheumatology. 2000;27:2743-2746
  18. 18. Rantapaa-Dahlqvist S, de Jong BA, Berglin E, Hallmans G, Wadell G, Stenlund H, et al. Antibodies against cyclic citrullinated peptide and IgA rheumatoid factor predict the development ofrheumatoid arthritis. Arthritis and Rheumatism. 2003;48:2741-2749
  19. 19. Nielen MM, van Schaardenburg D, Reesink HW, van, de Stadt RJ, van, der Horst-Bruinsma IE, de Koning MH, et al. Specific autoantibodies precede the symptoms of rheumatoid arthritis: A study of serial measurements in blood donors. Arthritis and Rheumatism. 2004;50:380-386
  20. 20. Nielen MM, van Schaardenburg D, Reesink HW, Twisk JW, van, de Stadt RJ, van, der Horst-Bruinsma IE, et al. Increased levels of C-reactive protein in serum from blood donors before the onset of rheumatoid arthritis. Arthritis and Rheumatism. 2004;50:2423-2427
  21. 21. Jorgensen KT, Wiik A, Pedersen M, Hedegaard CJ, Vestergaard BF, Gislefoss RE, et al. Cytokines, autoantibodies and viral antibodies in premorbid and postdiagnostic sera from patients with rheumatoid arthritis: Case-control study nested in a cohort of Norwegian blood donors. Annals of the Rheumatic Diseases. 2008;67:860-866
  22. 22. Shadick NA, Cook NR, Karlson EW, Ridker PM, Maher NE, Manson JE, et al. C-reactive protein in the prediction of rheumatoid arthritis in women. Archives of Internal Medicine. 2006;166:2490-2494
  23. 23. Kokkonen H, Soderstrom I, Rocklov J, Hallmans G, Lejon K, Rantapaa Dahlqvist S. Up-regulation of cytokines and chemokines predates the onset of rheumatoid arthritis. Arthritis and Rheumatism. 2010;62:383-391
  24. 24. Karlson EW, Chibnik LB, Tworoger SS, Lee IM, Buring JE, Shadick NA, et al. Biomarkers of inflammation and development of rheumatoid arthritis in women from two prospective cohort studies. Arthritis and Rheumatism. 2009;60:641-652
  25. 25. Waaler E. On the occurrence of a factor in human serum activating the specific agglutination of sheep red corpuscles. Acta Pathologica et Microbiologica Scandinavica. 1940;17:172-188
  26. 26. Nell VP, Machold KP, Stamm TA, Eberl G, Heinzl H, Uffmann M, et al. Autoantibody profiling as early diagnostic and prognostic tool for rheumatoid arthritis. Annals of the Rheumatic Diseases. 2005;64:1731-1736
  27. 27. Roberts-Thomson PJ, Wernick RM, Ziff M. Quantitation of rheumatoid factor by laser nephelometry. Rheumatology International. 1982;2:17-20
  28. 28. Jo’nsson T, Steinsson K, Jo’nsson H, Geirsson AJ, Thorsteinsson J, Valdimarsson H. Combined elevation of IgM and IgA rheumatoid factor has high diagnostic specificity for rheumatoid arthritis. Rheumatology International. 1998;18:119-122
  29. 29. Jo’nsson T, Arinbjarnarson S, Thorsteinsson J, Steinsson K, Geirsson AJ, Jo’nsson H, et al. Raised IgA rheumatoid factor (RF) but not IgM RF or IgG RF is associated with extra-articular manifestations in rheumatoid arthritis. Scandinavian Journal of Rheumatology. 1995;24:372-375
  30. 30. Jorgensen C, Legouffe MC, Bologna C, Brochier J, Sany J. IgA isotype rheumatoid factor in rheumatoid arthritis: Clinical implications. Clinical and Experimental Rheumatology. 1996;14:301-304
  31. 31. Pai S, Päi L, Birkenfeldt R. Correlation of serum IgA rheumatoid factor levels with disease severity in rheumatoid arthritis. Scandinavian Journal of Rheumatology. 1998;27:252-256
  32. 32. Van Snick JL, Van Roost E, Markowetz B, Cambiaso CL, Masson PL. Enhancement by IgM rheumatoid factor of in vitro ingestion by macrophages and in vivo clearance of aggregated IgG or antigen-antibody complexes. European Journal of Immunology. 1978;8:279-285
  33. 33. Brown PB, Nardella FA, Mannik M. Human complement activation by self-associated IgG rheumatoid factors. Arthritis and Rheumatism. 1982;25:1101-1107
  34. 34. Tighe H, Chen PP, Tucker R, Kipps TJ, Roudier J, Jirik FR, et al. Function of B cells expressing a human immunoglobulin M rheumatoid factor autoantibody in transgenic mice. The Journal of Experimental Medicine. 1993;177:109-118
  35. 35. Firestein GS. Rheumatoid synovitis and pannus. In: Hochberg MC, Silman AJ, Smolen JS, Weinblatt ME, Weisman MH, editors. Rheumatology. 3rd ed. Philadelphia, USA: Mosby; 2003. pp. 855-884
  36. 36. Schmidt D, Goronzy JJ, Weyand CM. CD4+ CD7− CD28− T cells are expanded in rheumatoid arthritis and are characterized by autoreactivity. The Journal of Clinical Investigation. 1996;97:2027-2037
  37. 37. Ziff M. Role of endothelium in the pathogenesis of rheumatoid synovitis. International Journal of Tissue Reactions. 1993;15:135-137
  38. 38. Roosnek E, Lanzavecchia A. Efficient and selective presentation of antigen-antibody complexes by rheumatoid factor B cells. The Journal of Experimental Medicine. 1991;173:487-489
  39. 39. Sebbag M, Chapuy-Regaud S, Auger I, et al. Clinical and pathophysiological significance of the autoimmune response to citrullinated proteins in rheumatoid arthritis. Joint, Bone, Spine. 2004;71(6):493-502
  40. 40. Young BJ, Mallya RK, Leslie RD. Anti-keratin antibodies in rheumatoid arthritis. British Medical Journal. 1979;2(6182):97-99
  41. 41. Sokolove J, Bromberg R, Deane KD, et al. Autoantibody epitope spreading in the pre-clinical phase predicts progression to rheumatoid arthritis. PLoS One. 2012;7:5
  42. 42. Goeb V, Jouen F, Gilbert D, Le Loet X, Tron F, Vittecoq O. Diagnostic and prognostic usefulness of antibodies to citrullinated peptides. Joint, Bone, Spine. 2009;76(4):343-349
  43. 43. Alenius GM. Psoriatic arthritis-new insights give new options for treatment. Current Medicinal Chemistry. 2007;14(3):359-366
  44. 44. Hendler A, Mulli TK, Hughes FJ, et al. Involvement of autoimmunity in the pathogenesis of aggressive periodontitis. Journal of Dental Research. 2010;89:1389-1394
  45. 45. Strollo R, Ponchel F, Malmstrom V, et al. Autoantibodies to posttranslationally modified type II collagen as potential biomarkers for rheumatoid arthritis. Arthritis and Rheumatism. 2013;65(7):1702-1712
  46. 46. Cruyssen BV, Cantaert T, Nogueira L, et al. Diagnostic value of anti-human citrullinated fibrinogen ELISA and comparison with four other anti-citrullinated protein assays. Arthritis Research & Therapy. 2006;8(4):R122
  47. 47. Chapuy-Regaud S, Nogueira L, Clavel C, Sebbag M, Vincent C, Serre G. IgG subclass distribution of the rheumatoid arthritis-specific autoantibodies to citrullinated fibrin. Clinical & Experimental Immunology. 2005;139(3):542-550
  48. 48. Masson-Bessi’ere C, Sebbag M, Girbal-Neuhauser E, et al. The major synovial targets of the rheumatoid arthritis-specific antifilaggrin autoantibodies are deiminated forms of the 𝛼-and 𝛽-chains of fibrin. The Journal of Immunology. 2001;166(6):4177-4184
  49. 49. Sebbag M, Moinard N, Auger I, et al. Epitopes of human fibrin recognized by the rheumatoid arthritis-specific autoantibodies to citrullinated proteins. European Journal of Immunology. 2006;36(8):2250-2263
  50. 50. Verpoort KN, Jol-van der Zijde CM, Papendrecht-van der Voort EA, et al. Isotype distribution of anti-cyclic citrullinated peptide antibodies in undifferentiated arthritis and rheumatoid arthritis reflects an ongoing immune response. Arthritis and Rheumatism. 2006;54(12):3799-3808
  51. 51. van Beers JJ, Schwarte CM, Stammen-Vogelzangs J, et al. The rheumatoid arthritis synovial fluid citrullinome reveals novel citrullinated epitopes in apolipoprotein E, myeloid nuclear differentiation antigen, and beta-actin. Arthritis and Rheumatism. 2013;65(1):69-80
  52. 52. Kuhn KA, Kulik L, Tomooka B, et al. Antibodies against citrullinated proteins enhance tissue injury in experimental autoimmune arthritis. The Journal of Clinical Investigation. 2006;116(4):961-973
  53. 53. Scherer HU, van, der Woude D, Ioan-Facsinay A, et al. Glycan profiling of anti-citrullinated protein antibodies isolated from human serum and synovial fluid. Arthritis and Rheumatism. 2010;62(6):1620-1629
  54. 54. Harre U, Lang SC, Pfeifle R, et al. Glycosylation of immunoglobulin G determines osteoclast differentiation and bone loss. Nature Communications. 2015;6:6651
  55. 55. Kelley WN. Textbook of Rheumatology. 5th ed. Philadelphia: W.B. Saunders; 1997. pp. 626-685
  56. 56. Kuettner KE, Aydelotte MB, Thonar EJ. Articular cartilage matrix and structure: A minireview. The Journal of Rheumatology. Supplement. 1991;27:46-48
  57. 57. Dodge GR, Pidoux I, Poole AR. The degradation of type II collagen in rheumatoid arthritis: An immunoelectron microscopic study. Matrix. 1991;11:330-338
  58. 58. Morgan K. What do anti-collagen antibodies mean? Annals of the Rheumatic Diseases. 1990;49:62-65
  59. 59. Kim WU, Yoo WH, Park W, Kang YM, Kim SI, Park JH, et al. IgG antibodies to type II collagen reflect inflammatory activity in patients with rheumatoid arthritis. The Journal of Rheumatology. 2000;27:575-581
  60. 60. Mullazehi M, Mathsson L, Lampa J, Ronnelid J. High anti-collagen type-II antibody levels and induction of proinflammatory cytokines by anti-collagen antibody-containing immune complexes in vitro characterise a distinct rheumatoid arthritis phenotype associated with acute inflammation at the time of disease onset. Annals of the Rheumatic Diseases. 2007;66:537-541
  61. 61. Lindh I, Snir O, Lönnblom E, Uysal H, Andersson I, Nandakumar KS, et al. Type II collagen antibody response is enriched in the synovial fluid of rheumatoid joints and directed to the same major epitopes as in collagen induced arthritis in primates and mice. Arthritis Research & Therapy. 2014;16:R143
  62. 62. Morgan K, Clague RB, Collins I, Ayad S, Phinn SD, Holt PJ. Incidence of antibodies to native and denatured cartilage collagens (types II, IX, and XI) and to type I collagen in rheumatoid arthritis. Annals of the Rheumatic Diseases. 1987;46:902-907
  63. 63. Mullazehi M, Mathsson L, Lampa J, Ronnelid J. Surface-bound anti-type II collagen-containing immune complexes induce production of tumor necrosis factor alpha, interleukin-1beta, and interleukin-8 from peripheral blood monocytes via Fc gamma receptor IIA: A potential pathophysiologic mechanism for humoral anti-type II collagen immunity in arthritis. Arthritis and Rheumatism. 2006;54:1759-1771
  64. 64. Courtenay JS, Dallman MJ, Dayan AD, Martin A, Mosedale B. Immunisation against heterologous type II collagen induces arthritis in mice. Nature. 1980;283:666-668
  65. 65. Wooley PH, Luthra HS, Stuart JM, David CS. Type II collagen-induced arthritis in mice. I. Major histocompatibility complex (I region) linkage and antibody correlates. The Journal of Experimental Medicine. 1981;154:688-700
  66. 66. Svensson L, Jirholt J, Holmdahl R, Jansson L. B cell-deficient mice do not develop type II collagen-induced arthritis (CIA). Clinical and Experimental Immunology. 1998;111:521-526
  67. 67. Wang Y, Kristan J, Hao L, Lenkoski CS, Shen Y, Matis LA. A role for complement in antibody-mediated inflammation: C5-deficient DBA/1 mice are resistant to collagen-induced arthritis. Journal of Immunology. 2000;164:4340-4347
  68. 68. Burkhardt H, Koller T, Engström A, Nandakumar KS, Turnay J, Kraetsch HG, et al. Epitope-specific recognition of type II collagen by rheumatoid arthritis antibodies is shared with recognition by antibodies that are arthritogenic in collagen-induced arthritis in the mouse. Arthritis and Rheumatism. 2002;46:2339-2348
  69. 69. Kraetsch HG, Unger C, Wernhoff P, Schneider C, Kalden JR, Holmdahl R, et al. Cartilage-specific autoimmunity in rheumatoid arthritis: Characterization of a triple helical B cell epitope in the integrin-binding-domain of collagen type II. European Journal of Immunology. 2001;31:1666-1673
  70. 70. Nandakumar KS, Bajtner E, Hill L, Böhm B, Rowley MJ, Burkhardt H, et al. Arthritogenic antibodies specific for a major type II collagen triple-helical epitope bind and destabilize cartilage independent of inflammation. Arthritis and Rheumatism. 2008;58:184-196
  71. 71. Nandakumar KS, Svensson L, Holmdahl R. Collagen type II-specific monoclonal antibody-induced arthritis in mice: Description of the disease and the influence of age, sex, and genes. The American Journal of Pathology. 2003;163:1827-1837
  72. 72. Cook AD, Rowley MJ, Mackay IR, Gough A, Emery P. Antibodies to type II collagen in early rheumatoid arthritis. Correlation with disease progression. Arthritis and Rheumatism. 1996;39:1720-1727
  73. 73. Burkhardt H, Sehnert B, Bockermann R, Engström A, Kalden JR, Holmdahl R. Humoral immune response to citrullinated collagen type II determinants in early rheumatoid arthritis. European Journal of Immunology. 2005;35:1643-1652
  74. 74. Uysal H, Nandakumar KS, Kessel C, Haag S, Carlsen S, Burkhardt H, et al. Antibodies to citrullinated proteins: Molecular interactions and arthritogenicity. Immunological Reviews. 2010;233:9-33
  75. 75. Brennan FM, McInnes IB. Evidence that cytokines play a role in rheumatoid arthritis. The Journal of Clinical Investigation. 2008;118(11):3537-3545
  76. 76. Dayer JM, Beutler B, Cerami A. Cachectin/tumor necrosis factor stimulates collagenase and prostaglandin E2 production by human synovial cells and dermal fibroblasts. The Journal of Experimental Medicine. 1985;162:2163-2168
  77. 77. Bertolini DR, Nedwin GE, Bringman TS, Smith DD, Mundy GR. Stimulation of bone resorption and inhibition of bone formation in vitro by human tumour necrosis factors. Nature. 1986;319:516-518
  78. 78. Buchan G et al. Detection of activated T cell products in the rheumatoid joint using cDNA probes to Interleukin-2 (IL-2) IL-2 receptor and IFN-gamma. Clinical and Experimental Immunology. 1988;71:295-301
  79. 79. Hirano T et al. Excessive production of interleukin 6/B cell stimulatory factor-2 in rheumatoid arthritis. European Journal of Immunology. 1988;18:1797-1801
  80. 80. Haworth C et al. Expression of granulocytemacrophage colony-stimulating factor in rheumatoid arthritis: Regulation by tumor necrosis factoralpha. European Journal of Immunology. 1991;21:2575-2579
  81. 81. Brennan FM et al. Detection of interleukin 8 biological activity in synovial fluids from patients with rheumatoid arthritis and production of interleukin 8 mRNA by isolated synovial cells. European Journal of Immunology. 1990;20:2141-2144
  82. 82. Brennan FM, Chantry D, Jackson A, Maini R, Feldmann M. Inhibitory effect of TNF alpha antibodies on synovial cell interleukin-1 production in rheumatoid arthritis. Lancet. 1989;2:244-247
  83. 83. Brennan FM, Chantry D, Jackson AM, Maini RN, Feldmann M. Cytokine production in culture by cells isolated from the synovial membrane. Journal of Autoimmunity. 1989;2(Suppl):177-186
  84. 84. Kavanaugh AF, Schulze-Koops H, Davis LS, Lipsky PE. Repeat treatment of rheumatoid arthritis patients with a murine anti-intercellular adhesion molecule 1 monoclonal antibody. Arthritis and Rheumatism. 1997;40:849-853
  85. 85. Deon D, Ahmed S, Tai K, et al. Cross-talk between IL-1 and IL-6 signaling pathways in rheumatoid arthritis synovial fibroblasts. Journal of Immunology. 2001;167:5395-5403
  86. 86. Elliott MJ et al. Treatment of rheumatoid arthritis with chimeric monoclonal antibodies to tumor necrosis factor alpha. Arthritis and Rheumatism. 1993;36:1681-1690
  87. 87. Elliott MJ et al. Randomised double-blind comparison of chimeric monoclonal antibody to tumour necrosis factor alpha (cA2) versus placebo in rheumatoid arthritis. Lancet. 1994;344:1105-1110
  88. 88. Maini RN et al. Randomized placebo controlled trial of multiple intravenous infusions of anti-TNFα monoclonal antibody with or without weekly methotrexate in rheumatoid arthritis. Arthritis and Rheumatism. 1998;41:1552-1563
  89. 89. Lipsky PE et al. 102 week clinical and radiological results from the ATTRACT trial: A 2 year, randomized, controlled, phase 3 trial of infliximab (Remicade) in patients with active rheumatoid arthritis despite methotrexate. Arthritis and Rheumatism. 2000;47(Suppl):S242
  90. 90. Feldmann M, Maini RN. Anti-TNFα therapy or rheumatoid arthritis: What have we learned? Annual Review of Immunology. 2001;19:163-196
  91. 91. Kay J, Calabrese L. The role of interleukin-1 in the pathogenesis of rheumatoid arthritis. Rheumatology. 2004;43(Suppl. 3):iii2-iii9
  92. 92. Chomarat P, Vannier E, Dechanet J, et al. Balance of IL-1 receptor antagonist/IL-1 in rheumatoid synovium and its regulation by IL-4 and IL-10. Journal of Immunology. 1995;154:1432-1439
  93. 93. Fontana A, Hengartner H, Weber E, Fehr K, Grob PJ, Cohen G. Interleukin 1 activity in the synovial fluid of patients with rheumatoid arthritis. Rheumatology International. 1982;2:49-53
  94. 94. Weckmann AN, Alcocer-Varela J. Cytokine inhibitors in autoimmune disease. Seminars in Arthritis and Rheumatism. 1996;26:539-557
  95. 95. Akira S, Taga T, Kishimoto T. Interleukin-6 in biology and medicine. Advances in Immunology. 1993;54:1-78
  96. 96. Kishimoto T. Interleukin-6: From basic science to medicine-40 years in immunology. Annual Review of Immunology. 2005;23:1-21
  97. 97. Muraguchi A, Hirano T, Tang B, Matsuda T, Horii Y, Nakajima K, et al. The essential role of B cell stimulatory factor 2 (BSF-2/IL-6) for the terminal differentiation of B cells. The Journal of Experimental Medicine. 1988;167:332-344
  98. 98. Jogo G, Bataille R, Pellat-Deceunynck C. Interleukin-6 is a growth factor for nonmalignant human plasmablasts. Blood. 2001;97:1817-1822
  99. 99. Dienz O, Eaton SM, Bond JP, Neveu W, Moquin D, Noubade R, et al. The induction of antibody production by IL-6 is indirectly mediated by IL-21 produced by CD4 T cells. The Journal of Experimental Medicine. 2009;206:69-78
  100. 100. Dayer JM, Choy E. Therapeutic targets in rheumatoid arthritis: The interleukin-6 receptor. Rheumatology. 2010;49:15-24
  101. 101. Lally F, Smith E, Filer A, et al. A novel mechanism of neutrophil recruitment in a coculture model of the rheumatoid synovium. Arthritis and Rheumatism. 2005;52:3460-3469
  102. 102. Teitelbaum SL. Bone resorption by osteoclasts. Science. 2000;289:1504-1508
  103. 103. Schett G. Cells of the synovium in rheumatoid arthritis. Osteoclasts. Arthritis Research & Therapy. 2007;9:203
  104. 104. Paleolog EM. Angiogenesis in rheumatoid arthritis. Arthritis Research. 2002;4:S81-S90
  105. 105. Keck PJ, Hauser SD, Krivi G, Sanzo K, Warren T, Feder J, et al. Vascular permeability factor, an endothelial cell mitogen related to PDGF. Science. 1989;246:1309-1312
  106. 106. Ballara S, Taylor PC, Reusch P, Malme D, Feldmann M, Maini RN, et al. Raised serum vascular endothelial growth factor levels are associated with destructive change in inflammatory arthritis. Arthritis and Rheumatism. 2001;44:20552064
  107. 107. Nakahara H, Song J, Sugimoto M, Hagihara K, Kishimoto T, Yoshizaki K, et al. Anti-interleukin-6 receptor antibody therapy reduces vascular endothelial growth factor production in rheumatoid arthritis. Arthritis and Rheumatism. 2003;48:1521-1529
  108. 108. Choy EH, Isenberg DA, Garrood T, Farrow S, Ioannou Y, Bird H, et al. Therapeutic benefit of blocking interleukin-6 activity with an anti-interleukin-6 receptor monoclonal antibody in rheumatoid arthritis: A randomized, double-blind, placebo-controlled, dose-escalation trial. Arthritis and Rheumatism. 2002;46:3143-3150
  109. 109. Nishimoto N, Yoshizaki K, Miyasaka N, Yamamoto K, Kawai K, Takeuchi T, et al. Treatment of rheumatoid arthritis with humanized anti-interleukin-6 receptor antibody: A multicenter, double-blind, placebo-controlled trial. Arthritis and Rheumatism. 2004;50:1761-1769
  110. 110. Maini RN, Taylor PC, Szechinski J, Pavelka K, Broll J, Balint G, et al. Double-blind randomized controlled clinical trial of the interleukin-6 receptor antagonist, tocilizumab, in European patients with rheumatoid arthritis who had an incomplete response to methotrexate. Arthritis and Rheumatism. 2006;54:2817-2829
  111. 111. Smolen JS, Beaulieu A, Rubbert-Roth A, Ramos-Remus C, Rovensky J, Alecock E, 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
  112. 112. Genovese MC, McKay JD, Nasonov EL, Mysler EF, da Silva NA, Alecock E, 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 and Rheumatism. 2008;58:2968-2980
  113. 113. Emery P, Keystone E, Tony HP, Cantagrel A, van Vollenhoven R, Sanchez A, 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. Annals of the Rheumatic Diseases. 2008;67:1516-1523
  114. 114. Nishimoto N, Hashimoto J, Miyasaka N, Yamamoto K, Kawai S, Takeuchi T, et al. Study of active controlled monotherapy used for rheumatoid arthritis, an IL-6 inhibitor (SAMURAI): Evidence of clinical and radiographic benefit from an x ray reader-blinded randomised controlled trial of tocilizumab. Annals of the Rheumatic Diseases. 2007;66:1162-1167
  115. 115. Fleischmann R, Burgos-Vargas R, Ambs P, Alecock E, Kremer J. LITHE: Tocilizumab inhibits radiographic progression and improves physical function in rheumatoid arthritis (RA) patients (Pts) at 2 Yrs with increasing clinical efficacy over time. Arthritis and Rheumatism. 2009;60(10):S238-S239
  116. 116. Caporali R, Idolazzi L, Bombardieri S. Tocilizumab in the treatment of patients with rheumatoid arthritis in real clinical practice: Results of an Italian observational study. Clinical and Experimental Rheumatology. 2017;35(6):919-928
  117. 117. Rubbert-Roth A, Furst DE, Nebesky JM, Jin A, Berber E. A review of recent advances using tocilizumab in the treatment of rheumatic diseases. Rheumatology and Therapy. 2018;5(1):21-24

Written By

Mohd Wajid Ali Khan and Wahid Ali Khan

Submitted: 27 March 2018 Reviewed: 26 October 2018 Published: 22 November 2018