GET THE APP

Targeting of Interleukin-6 for the Treatment of Rheumatoid Arthri
Rheumatology: Current Research

Rheumatology: Current Research
Open Access

ISSN: 2161-1149 (Printed)

+44-20-4587-4809

Review Article - (2014) Volume 0, Issue 0

Targeting of Interleukin-6 for the Treatment of Rheumatoid Arthritis: A Review and Update

Toshio Tanaka1,2*, Atsushi Ogata2,3 and Tadamitsu Kishimoto4
1Department of Clinical Application of Biologics, Osaka University Graduate School of Medicine, Osaka University, Osaka, Japan
2Department of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
3Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Osaka University Graduate School of Medicine, Osaka University, Osaka, Japan
4Laboratory of Immunoregulation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
*Corresponding Author: Toshio Tanaka, Department of Clinical Application of Biologics, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita City, Osaka 565-0871, Japan, Tel: +81-6-6879-3838, Fax: +81-6-6879-3839 Email:

Abstract

Rheumatoid Arthritis (RA) is a chronic inflammatory disease, characterized by persistent joint inflammation, systemic inflammation and immunological abnormalities. Since IL-6 plays a major role in the development of these characteristics, its targeting could reasonably be expected to constitute a novel therapeutic strategy for the treatment of RA. Tocilizumab, a humanized anti-human IL-6 receptor monoclonal antibody, has demonstrated its outstanding clinical efficacy and tolerable safety profile in phase III clinical trials for RA patients, resulting in its worldwide approval for moderate-to-severe active RA. Post-marketing clinical trials have confirmed its efficacy. This success led to the development of various other IL-6 inhibitors. Further clinical studies including head-to-head comparative studies, and clarification of the mechanisms through which tocilizumab exerts its clinical effects can be expected to identify RA patients who should be treated with tocilizumab as a first-line biologic. In addition, recent case reports and pilot studies indicate that therapies targeting IL-6 will be widely applicable to the treatment of various intractable immune-mediated diseases.

<

Keywords: Rheumatoid arthritis; Interleukin-6; A humanized antiinterleukin- 6 receptor antibody; Tocilizumab

Introduction

Rheumatoid Arthritis (RA), a chronic disease affecting 0.5-1% of adults, is characterized by persistent synovitis, systemic inflammation and immunological abnormalities. Uncontrolled active RA causes joint damage, disability, diminished quality of life and cardiovascular and other comorbidities [1,2]. Interleukin 6 (IL-6), a typical cytokine featuring redundancy and pleiotropic activity, plays a key role in the development of RA [3-7]. IL-6 promotes the imbalance between Th17 cells and regulatory T cells (Treg) and the production of autoantibodies such as Rheumatoid Factor (RF) and Anti-Citrullinated Peptide Antibody (ACPA). It also promotes synovial inflammation and cartilage and bone destruction as well as systemic features including cardiovascular, psychological and skeletal disorders. In this review article, we highlight the rationale for strategies targeting IL-6 for the treatment of RA, current evidence of efficacy and safety of tocilizumab, a humanized anti-IL-6 receptor antibody, and discuss its place in the biological treatment of RA and assess the possibility of its widespread use for the treatment of various other immune-mediated diseases.

The Pathological Role of IL-6 in RA

Biological activity of IL-6 related to pathogenesis of RA

IL-6 was originally cloned in 1986 as a B cell stimulatory factor 2 that induces activated B cells to produce immunoglobulin [8]. Subsequent in vitro studies using recombinant IL-6 have demonstrated that IL-6 is a multifunctional cytokine [3,9,10]. RA is characterized by synovial inflammation and hyperplasia, immunological abnormalities leading to autoantibody production such as RF and ACPA, cartilage and bone destruction and systemic features [1,2]. Although the precise cause of RA remains unknown, it has been hypothesized that a multistep progression is required for the development of RA [2]. For the first step, environment-gene interactions promote loss of tolerance to self-proteins that contain a citrulline residue. This is followed by localization of the inflammatory response in the joints and synovitis, which is initiated and perpetuated by positive feedback loops, and in turn promotes systemic disorders. IL-6 appears to play a key role in all these steps in the progression of RA [4-7].

IL-6 contributes to the production of autoantibodies such as RF or ACPA by acting on plasmablasts [11]. Alternatively, IL-6 in the presence of transforming growth factor (TGF)-β promotes Th17 differentiation but inhibits TGF-β-induced Treg from CD4+ naïve T cells [12,13], leading to imbalance between Th17 and Treg. This imbalance is recently considered to contribute to the development of immunological abnormalities in RA [1,2,4-7].

Systemic inflammatory symptoms, signs and laboratory findings related to RA include fever, malaise, sleep disturbance, muscle weakness, anemia, thrombosis, C-reactive protein (CRP) elevation, hypercoagulability and hypoalbuminemia, and these are mostly mediated by IL-6. When IL-6 acts on hepatocytes, it induces a broad spectrum of acute phase proteins including CRP, Serum Amyloid A (SAA), haptoglobulin, antichymotrypsin, fibrinogen and hepcidin, whereas it reduces the production of albumin, fibronectin, transferrin and cytochrome p450 (CYP) [14,15]. High levels of hepcidin induced by IL-6 block iron transporter ferroportin 1 on macrophages, hepatocytes and gut epithelial cells, leading to hypoferremia and anemia associated with chronic inflammation [16]. Long-term high concentrations of SAA lead to amyloid A amyloidosis, a serious complication of RA, in which amyloid fibril deposition causes progressive deterioration in various organs [17]. RA patients often suffer from thrombocytosis, also mediated by IL-6, which promotes the differentiation of megakaryocytes into platelets [18]. Furthermore, IL-6 reduces the activity of CYP and CYP3A4 isozyme in particular. Simvastatin is a CYP3A4 substrate, so that clinicians should exercise caution when co-prescribing IL-6 inhibitors and CYP-metabolized drugs such as simvastatin [19]. Cardiovascular events are the major determinants of poor prognosis for RA [20] and increased IL-6 expression is associated with the development of atherosclerosis and cardiovascular diseases such as obesity, myocardial infarction and type II diabetes [21,22]. IL-6 is also an important messenger molecule that connects peripheral regulatory processes with the central nervous system and affects sleeprelated symptoms and fatigue, while changes in sleep duration and quality stimulate an increase in IL-6 concentrations in the circulation [23]. In addition, high levels of IL-6 reportedly increase the risk of muscle mass and strength loss [24,25].

IL-6 has been demonstrated to play a major role in local inflammation causing joint destruction by inducing endothelial cells to produce IL-8 and monocyte chemoattractant protein-1, and to activate expression of adhesion molecules and recruitment of leukocytes to involved joints [26,27]. Moreover, synoviocytes can produce IL-6 [28], while IL-6 can induce synoviocyte proliferation [29] and osteoclast differentiation through Receptor Activator of NF-kappa B Ligand (RANKL) expression [30,31]. This stimulation by IL-6 is also associated with the development of osteoporosis and fractures [32,33]. Enhanced angiogenesis and vascular permeability of synovial tissue are pathological features of RA resulting from the excess production of Vascular Endothelial Growth Factor (VEGF), which is induced by IL-6 in synovial fibroblasts [34,35]. Finally, IL-6 and IL-1 synergistically enhance the production of matrix metalloproteinases (MMPs) from synovial cells, which may lead to cartilage and joint destruction [36,37]. Therefore, IL-6 plays a key role in the induction of immunological abnormalities and in the development of joint and systemic inflammation of RA as well as its complications.

IL-6 expression and gene polymorphisms of IL-6 and IL-6 receptor in patients with RA

Elevated IL-6 levels were found in serum and synovial fluid of patients with RA [38-40]. These levels correlated with clinical symptoms such as morning stiffness, the number of involved joints and laboratory indices of disease activity [41-43]. Moreover, treatment with Disease-Modifying Antirheumatic Drugs (DMARDs) has been shown to be associated with reduced IL-6 serum concentrations and reduction of IL-6 during the first 12 months is reportedly a prognostic marker for clinical outcome [44].

Genetic polymorphisms (-622 or -174) in the IL-6 gene promoter, in which the -174 polymorphism was found to affect IL-6 levels [45], did not appear to increase susceptibility to RA [46]. However, a recent meta-analysis of nine studies regarding the association between IL-6 gene polymorphisms and RA showed that the -174 polymorphism might confer susceptibility to RA in Europeans [47]. Moreover, this polymorphism is reportedly associated with an increase in radiographic damage of joints in RA patients who were RF- or ACPA- positive [48] or with disease activity [49].

A single nucleotide polymorphism (rs8192284A>C) located at the proteolytic cleavage site of the IL-6 receptor (IL-6R) has been identified, which results in an amino acid substitution from aspartic acid to alanine (Asp358Ala) [50], while carriage of the C allele has been shown to correlate with higher levels of soluble IL-6R (sIL- 6R) in Japanese [51] and Caucasian populations [52]. Although the positive association of plasma sIL-6R concentrations with the number of C alleles in RA patients is generally agreed upon [53], the results regarding the association between this IL-6R polymorphism and RA appeared complicated. It was previously demonstrated that concentrations of sIL-6R as well as IL-6 were higher in the serum of RA patients than in healthy controls [40] and that synovial levels of sIL-6R and IL-6 were significantly higher in RA than in osteoarthritic patients and correlated with the severity of joint damage [30]. On the other hand, it was demonstrated that the C allele frequency was not higher but lower in RA patients than in controls, which suggests that lower sIL-6R levels could be a risk factor for RA [54]. Moreover, two recent articles reported that the polymorphism of IL-6R, which results in higher serum concentrations of sIL-6R, is associated with a lower risk of coronary heart disease [55,56]. This has led to the hypothesis that the point mutation leads to an increased cleavage of the transmembrane IL-6R from the cell surface of hepatocytes, monocytes and macrophages and consequently to less IL-6-induced signaling by these cells [57]. Another hypothesis is that the reduced risk of coronary heart disease is due to a more efficient buffering of secreted IL-6 by the sIL-6R/soluble gp130 system and consequently to lower overall IL-6 activity [58].

Findings from RA animal models

Various RA models have been used for the pathological analyses of RA and for determining therapeutic targets, and evidence in support of the key role of IL-6 in the development of RA has come from many studies of RA animal models.

Collagen-Induced Arthritis (CIA) is the most well-known animal model of RA, in which injection of mice with type II collagen produces an immune response directed at connective tissues. In the CIA model, IL-6 has been shown to perform a major role in the development and progression of joint destruction since the incidence and severity of arthritis is reduced in mice with IL-6 gene deficiency or IL-6 blockade by means of anti-IL-6R antibody (Ab) [59-61]. Moreover, immunization with type II collagen predominantly increased the frequency of Th17 cells in the CIA model, while treatment of the mice with anti-IL-6R Ab during the priming markedly suppressed the induction of Th17 cells and arthritis development. However, treatment with the Ab on day 14 failed to suppress Th17 differentiation and arthritis [62].

IL-6 gene knockout mice with Antigen-Induced Arthritis (AIA), an immune complex model of RA, did not develop joint swelling, while CD4+ T lymphocytes from these mice showed reduced antigeninduced proliferation and produced less IL-17 and RANKL [63,64].

SKG mice spontaneously develop autoimmune arthritis with ageing due to a spontaneous mutation in the zeta-chain-associated protein kinase-70 (ZAP-70) gene, resulting in autoreactive peripheral T cells that cause arthritis [65]. Synovial fluids of arthritic mice were found to contain large amounts of IL-6, while IL-6 gene deficiency completely inhibited the development of arthritis, and 20% of tumor necrosis factor (TNF)-α-deficient mice developed arthritis [66]. Moreover, IL- 6-deficient SKG mice completely lacked IL-17 producing CD4+ T cells, whereas TNF-α-deficient SKG mice possessed equivalent numbers of IL-17 producing CD4+ T cells as did intact SKG mice [67].

Human adult T cell leukemia virus (HTLV)-1 Tax transgenic mice constitute a genetically modified arthritis model and spontaneously develop arthritis [68]. IL-6 expression was found to be high in these mice [69], while IL-6 deficiency prevented the development of arthritis but TNF deficiency did not.

Knock-in mice, known as F759 mice, with substitution of a mutation at position tyrosine-759 for phenylalanine in IL-6 family cytokine receptor subunit gp130, which is a binding site of the src homology 2 domain-bearing protein tyrosine phosphatase (SHP)-2, spontaneously and age-dependently developed an RA-like joint disease featuring an increased number of autoreactive T cells and autoantibodies, indicating that constitutive activation of IL-6 signaling is involved in the development of RA-like features [70]. The IL-6 amplifier loop, which features an age-dependent increase in Th17 cells, excessive IL-6 production, and is a positive feedback loop for NF-κB signaling, as well as a signal transducer and activator of transcription (STAT)3 activation in non-immune cells, has been shown to be responsible for the development of arthritis in ageing F759 mice [71-73].

Arthritis of anti-type II collagen Ab-induced arthritis (CAIA) is another arthritis model but in this model the priming phase of T cell dependent Ab generation is skipped. Although IL-6 is also elevated in this model, arthritis is suppressed in TNF- but not IL-6-deficient mice, indicating that TNF plays a more significant role than IL-6 in joint inflammation in CAIA [74].

In a glucose-6-phosphate isomerase (GPI)-induced arthritis model, administration of anti-IL-6R Ab on day 0 or 3 suppressed Th17 differentiation and protected against arthritis induction while injection of the Ab on day 14, at the peak of arthritis, did not bring about any improvement in arthritis [75].

Stimulation of murine naïve CD4+ T cells with IL-6 and TGF-β, which are essential for the induction of Th17, induced a marked expression of the aryl hydrocarbon receptor (Ahr) [76], which is known as a dioxin receptor. Ahr is present in cytoplasm and, upon binding with a ligand, translocates to the nucleus and dimerizes with the Ahr nuclear translocator (Arnt). The resultant Ahr-Arnt complex then binds to specific sequences, designated as xenobiotic responsive elements and exerts a variety of biological effects [77]. The experiments demonstrated that Ahr interacts with and negatively regulates both STAT1 and STAT5 activation, thus leading to the augmentation of Th17 differentiation. As expected, Ahr deficiency in T cells showed a significant reduction in Th17 [76] and suppressed the development of collagen-induced arthritis [78], pointing to the importance of the pathological IL-6-Ahr-Th17 axis in the RA model.

These observations obtained from various models of RA clearly indicate that IL-6 is essential for the induction of immunological abnormalities and the development of arthritis and that the pathological role of IL-6 is different from that of TNF-α which predominantly acts on the development of joint inflammation.

Clinical Efficacy and Safety of Tocilizumab, a Humanized Anti-IL-6 Receptor Antibody for RA

The findings described in the preceding section have led to expectations that targeting of IL-6 could be a novel treatment strategy for RA. IL-6 transmits its signal through binding to transmembrane IL-6R or sIL-6R [3,79-81]. After binding of IL-6 to IL-6R, the resultant IL-6/transmembrane IL-6R complex or IL-6/sIL-6R complex associates with gp130 and induces homodimerization of gp130, which triggers a signal transduction system [82,83]. Tocilizumab (TCZ) is a humanized anti-human IL-6R monoclonal Ab of the IgG1 class that was generated by grafting the complementarity-determining regions of a mouse anti-human IL-6R Ab onto human IgG1 [84]. TCZ blocks IL- 6-mediated signal transduction through inhibition of IL-6 binding to transmembrane IL-6R and sIL-6R.

Phase I/II clinical trials

A randomized, double-blind, placebo-controlled, dose-escalation phase I trial was performed in the UK with 45 patients with active RA [85]. Patients were sequentially allocated to receive a single intravenous (IV) injection of 0.1, 1, 5 or 10 mg/kg of TCZ or placebo. At week 2, a significant difference was observed between the patients treated with 5 mg/kg of TCZ and with the placebo, with five patients (55.6%) in the TCZ cohort and none in the placebo cohort showing a 20% improvement according to the American College of Rheumatology (ACR) standards.

An open label phase I/II clinical study was performed in Japan with 15 patients with active RA [86]. Patients received IV administration of 3 doses (2, 4 or 8 mg/kg) of TCZ biweekly for 6 weeks with pharmacokinetic assessment. Safety and efficacy of the drug were evaluated after continuation of the TCZ treatment for 24 weeks. The treatment was found to be well tolerated at all doses with no severe adverse events (AEs). Although there was no statistically significant difference in efficacy among the three different dose groups, 9 of 15 patients (60%) achieved ACR20 response at week 6, while 86% achieved ACR20 and 33% ACR50 at week 24.

In a multicenter, double-blind, placebo-controlled trial, 164 patients with refractory RA were randomized to receive either TCZ (4 or 8 mg/kg) or placebo [87]. TCZ was administered IV three times every 4 weeks (q4w). After 12 weeks, ACR20 response was observed in 78%, 57% and 11% of RA patients treated with TCZ 8, 4 mg/kg and placebo, respectively.

The CHARISMA study was a phase II, double-blind, randomized, placebo-controlled, multicenter trial of TCZ with European RA patients who had showed an incomplete response to methotrexate (MTX) [88]. The 359 patients with active RA enrolled in this study were randomized to one of the following seven treatment arms: TCZ at doses of 2, 4 or 8 mg/kg, either as monotherapy or in combination with MTX, or MTX plus placebo. ACR20 response at week 16 was attained by 61% and 63% of the patients receiving 4 mg/kg and 8 mg/kg of TCZ as monotherapy, respectively, and by 63% and 74% of the patients receiving the same doses of TCZ plus MTX, respectively, compared with 41% of the patients receiving placebo plus MTX. Statistically significant ACR50 and ACR70 responses were observed in patients receiving combination therapy with either 4 mg/kg or 8 mg/kg of TCZ plus MTX.

Phase III clinical trials

Subsequent to phase I and II studies, seven phase III clinical trials of TCZ proved its efficacy either as monotherapy or in combination with MTX or other DMARDs for adult patients with moderate-tosevere active RA [89-97].

The SAMURAI study was the first phase III, double-blind, placebocontrolled, multicenter trial of TCZ to evaluate the ability of TCZ monotherapy to inhibit progression of structural joint damage [89]. The 306 patients with active RA of less than 5 years’ duration enrolled in this study were allocated to receive either TCZ monotherapy at 8 mg/ kg q4w or conventional DMARDs for 52 weeks. At week 52, the TCZ group showed statistically significantly less radiographic change in the van der Heijde-modified Total Sharp Score (mTSS) (mean 2.3, 95% Confidence Interval (CI): 1.5 to 3.2) than the DMARD group (mean 6.1, 95% CI: 4.2 to 8.0, p<0.01). Signs and symptoms also improved with TCZ monotherapy.

The phase III, double-blind, placebo-controlled, multicenter TOWARD study was conducted to observe the effect of TCZ added to a stable dosage of conventional DMARDs for moderate-to-severe active RA in 1,220 patients with an inadequate response to DMARDs [90]. Patients remained on stable doses of DMARDs and received TCZ 8 mg/kg or placebo q4w for 24 weeks. At week 24, the percentage of patients attaining ACR20 response was significantly higher for the TCZ plus DMARD group than for the placebo group (61% vs. 25%, p<0.0001).

The RADIATE study examined the efficacy and safety of TCZ for patients with RA refractory to TNF inhibitor therapy [91]. A total of 499 patients with an inadequate response to one or more TNF inhibitors were randomly assigned to receive 8 mg/kg, 4 mg/kg TCZ or placebo with stable MTX for 24 weeks. ACR20 response was attained at week 24 by 50.0% of the patients in the 8 mg/kg, 30.4% in the 4 mg/kg, and 10.1% in the control group. 28-joint disease activity score (DAS28) remission rates (DAS28<2.6) at week 24 were clearly dose-related, being attained by 30.1%, 7.6% and 1.6% of the 8 mg/kg, 4 mg/kg and control group, respectively.

The aim of the multicenter double-blind, randomized, placebocontrolled, parallel group phase III OPTION study was to observe the therapeutic effect of TCZ added to a stable dosage of MTX on moderate-to-severe active RA of patients with an inadequate response to MTX [92]. For this study, 623 patients were randomized to receive TCZ (4 or 8 mg/kg) or placebo q4w, with MTX continued at stable pre-study doses (10-25 mg/week). Rescue therapy with TCZ (8 mg/kg) was offered at week 16 to patients with less than 20% improvement in both swollen and tender joint counts. The primary endpoint was the proportion of patients with ACR20 response at week 24, which was achieved by 59%, 48% and 26% of the patients in the TCZ 8 mg/ kg, 4 mg/kg and control group, respectively. In the OPTION study, serum biochemical markers of bone formation (osteocalcin and N-terminal propeptide of type I collagen [PINP]), bone resorption (C-terminal crosslinking telopeptide of type I collagen [CTX-I] and CTX-I generated by matrix metalloproteinase [ICTP]), cartilage metabolism (N-terminal propeptide of type IIA collagen [PIIANP] and collagen helical peptide [HELIX-II]), and MMP-3 were measured [93]. At 4 weeks, TCZ had induced marked dose-dependent reductions in PIIANP, HELIX-II and MMP-3 levels, which were associated with enhancement of bone formation marker, PINP. TCZ also induced significant reductions in the bone degradation markers such as CTX-1 and ICTP, indicating the beneficial effect of TCZ on bone turnover.

The SATORI study investigated the clinical efficacy and safety of TCZ monotherapy for active RA patients with an inadequate response to low dose MTX [94]. For this study, 125 patients were allocated to receive TCZ 8 mg/kg q4w plus MTX placebo or TCZ placebo plus MTX 8 mg/week for 24 weeks. At week 24, the ACR20 response rate was 80.3% for the TCZ and 25.0% for the MTX group. In fact, the ACR response of the TCZ group was superior to that of the MTX group at all time points.

The LITHE trial confirmed the efficacy of TCZ for preventing the progression of joint destruction. This study was a three-arm, randomized double-blind, placebo-controlled study [95]. Patients were randomized to receive TCZ (4 or 8 mg/kg q4w) plus MTX or placebo plus MTX. Stepwise rescue therapy starting at week 16 was allowed if patients did not respond. At 1 year, patients treated with TCZ plus MTX showed less progression of joint damage, as evaluated with the Genant-modified Total Sharp Score (GmTSS) method (mean: 0.34 for TCZ 4 mg/kg plus MTX, 0.29 for TCZ 8 mg/kg plus MTX), than did those treated with MTX alone (1.13). Moreover, at 2 years, the mean change in GmTSS from baseline was significantly lower for patients initially randomized to TCZ 4 mg/kg plus MTX (0.58) or TCZ 8 mg/kg plus MTX (0.37) than for patients initially randomized to placebo plus MTX (1.96) [96].

The 24-week, double-blind, double-dummy, parallel-group AMBITION study was conducted to determine the efficacy of TCZ monotherapy vs. MTX for patients with active RA for whom previous treatment with MTX/biological agents had not failed [97]. For this trial, 673 patients were randomized to receive either TCZ 8 mg/kg q4w, MTX starting at 7.5 mg/week and titrated to 20 mg/week within 8 weeks or placebo for 8 weeks followed by TCZ 8 mg/kg. Results for TCZ were better than for MTX treatment with a higher ACR20 response (69.9% vs. 52.5%, p<0.001) and DAS28 remission rate (33.6% vs. 12.1%) at week 24.

The above trial data demonstrate the outstanding therapeutic efficacy of TCZ for adult patients with moderate-to-severe active RA. Outcomes attained in the seven phase III studies are summarized in Figure 1. TCZ has demonstrated its superior efficacy for the treatment of RA, either as monotherapy or in combination with MTX or other DMARDs, in comparison with control groups. As a result, TCZ has now been approved for the treatment of RA in more than 100 countries worldwide [4-7]. The recommended posology of TCZ is 8 mg/kg q4w in Japan and the EU. In the United States, the recommended starting dose is 4 mg/kg q4w followed by an increase to 8 mg/kg depending on clinical response.

rheumatology-current-antirheumatic-drugs

Figure 1: The efficacy of tocilizumab in seven phase III trials. Tocilizumab as monotherapy or in combination with disease modifying antirheumatic drugs (DMARDs) or methotrexate (MTX) has shown superior efficacy compared with control in the treatment of RA. The percentages of ACR20, 50, 70 and remission responses evaluated at week 52 in SAMURAI and LITHE trials or at week 24 in other clinical trials are shown. TCZ: tocilizumab; IR: inadequate response.

Phase IIIb and IV clinical trials and clinical practice

In addition, the efficacy of TCZ was confirmed in subsequent clinical trials and actual medical practice.

The Rose study showed rapid improvement in clinical outcomes [98]. It was a 24-week, randomized, double-blind, placebo-controlled, parallel-group, multicenter phase IIIb clinical trial. Patients were randomly assigned at a 2:1 ratio to administration of TCZ 8 mg/ kg (n = 412) or placebo (n = 207) while background administration of DMARDs was continued for both groups. The primary efficacy endpoint, that is, the percentage of patients achieving ACR50 response at week 24, was higher for the TCZ than for the placebo group (30.1% vs. 11.2%, p<0.0001). A substudy examining early response to therapy showed improvement in patients’ global assessment of disease activity, pain and DAS28 for treatment with TCZ by day 7.

The ACT-SURE trial was a phase IIIb, open-label, single-arm, 6-month study, which was conducted with a patient population resembling one to be expected in clinical practice [99]. Patients, who were categorized as TNF inhibitor-naïve (never received TNF inhibitor), TNF inhibitor-previous (TNF inhibitor therapy discontinued for more than 2 months) or TNF inhibitor-recent (TNF inhibitor discontinued for less than 2 months), received open-label TCZ 8 mg/kg q4w with or without DMARDs for 24 weeks. A total of 1,681 patients (976 TNF inhibitor-naïve, 298 TNF inhibitor-previous and 407 TNF inhibitorrecent) were treated. Of the TNF inhibitor-naïve patients, 70.5% had attained ACR20, 51.9% ACR50 and 31.8% ACR70 responses at week 24, while for the patients with TNF inhibitor-previous the corresponding findings were 60.7%, 35.2% and 17.8%, and 62.7%, 42.3% and 19.7% for those with TNF inhibitor-recent.

The ACT-STAR study was a 24-week, prospective, open-label study conducted in the USA [100]. Of the 886 patients enrolled, 163 were assigned to monotherapy with TCZ 8 mg/kg. The remaining 723 patients were assigned to receive combination therapy, with 363 randomized to receive a starting dose of TCZ 4 mg/kg plus DMARDs and 360 a starting dose of TCZ 8 mg/kg plus DMARDs. The dosage for 152 patients (41.9%) of the initial TCZ 4 mg/kg plus DMARDs group was increased to TCZ 8 mg/kg plus DMARDs at week 8 and for 68 patients (18.7%) after 8 week. Overall, the 24-week study was completed by 82.5% of patients. By week 24, 49.7% of the patients in the 8 mg/kg TCZ plus DMARDs group had attained ACR20, 27.1% ACR50 and 10.3% ACR70, while the corresponding values for the patients in the 8 mg/kg TCZ monotherapy group were 47.9%, 24.5% and 7.4%. The efficacy of TCZ monotherapy and combination therapy with DMARDs was thus similar, while the safety profiles for the two groups were also similar.

For the TAMARA study conducted in Germany, 286 patients, 41.6% of whom had been previously treated with TNF inhibitors, were registered to determine the effectiveness and safety of TCZ [101,102]. Of the intention-to-treat patients, 57% attained the primary end point of low DAS (DAS<3.2), 47.6% showed DAS remission and a European League Against Rheumatism (EULAR) good response was attained by 54.9%, while ACR50/70 response rates at week 24 were 50.7% and 33.9%, respectively. Remission rates determined with the new ACR/ EULAR Boolean-based criteria for clinical studies were 15.0% after 12 weeks and 20.3% after 24 weeks, while the clinical disease activity index (CDAI) and simplified disease activity index (SDAI) remission rates were 24.1% and 25.2%, respectively.

In the nationwide registry of biological therapies established in Denmark, DANBIO, 178 patients with RA treated with TCZ have been identified, 93% of whom had previously received one or more TNF inhibitors [103]. The disease activity in these patients decreased at all time points, with DAS28 remission rates of 39% for TCZ treatment after 24 weeks and 58% after 48 weeks. EULAR good or moderate response rates were 88% at week 24 and 84% at week 48. These response rates were comparable to those for patients switching to their second TNF inhibitors as well as to those previously observed in phase III clinical trials.

In Japan, 229 patients were registered in the REACTION study to evaluate the efficacy and tolerability of TCZ in daily clinical practice for RA patients in Japan [104,105]. In this study, 55% of the patients received MTX concomitantly with TCZ and 63% had previously received anti-TNF treatment. Average DAS28 significantly decreased from 5.70 to 3.25 after 24 weeks of therapy. EULAR good response and DAS 28 remission response was attained after 24 weeks by 57.4% and 40.7% of the patients, respectively. After 52 weeks, clinical remission was observed in 43.7% of the patients, radiographic non-progression in 62.8% and functional remission in 26.4%. The retention rates at 24 and 52 weeks were 79.5% and 71.1%, respectively.

The ACT-RAY trial compared TCZ plus MTX therapy with TCZ monotherapy in a setting that closely resembled real-life clinical practice [106]. The trial was a double-blind, 2-year study in which patients whose RA had remained active in spite of MTX administration were randomly assigned to either continue MTX with the addition of TCZ 8 mg/kg q4w or switch to TCZ plus placebo. Of the 556 randomly assigned patients, 512 (92%) completed 24 weeks of the trial. ACR20/50/70 response rates were 71.5%, 45.5% and 24.5% for the TCZ plus MTX group and 70.3%, 40.2% and 25.4% for the TCZ plus placebo group, respectively, showing that TCZ plus MTX combination therapy was clinically not superior to TCZ monotherapy.

The Effect of tocilizumab on extra-articular symptoms and complications of RA

As described elsewhere, IL-6 also plays a role in the development of systemic inflammatory symptoms and complications related to RA, so that TCZ treatment was expected to ameliorate such symptoms and/or prevent the development of the complications.

Assessment of functional disability according to the Health Assessment Questionnaire Disability Index (HAQ-DI) [95,96,107] or of health status according to the Arthritis Impact Measurement Scale 2 (AIMS-2) and Short Form-36 (SF-36) showed improvements in patients with RA treated with TCZ [108]. Moreover, sleep quality assessed according to the Pittsburgh Sleep Quality Index (PSQI) improved while daytime sleepiness measured on the Epworth Sleepiness Scale and fatigue according to the Functional Assessment of Chronic Illness Therapy Fatigue Scale (FACIT-FFS) significantly decreased after TCZ treatment [109].

Increased production of hepcidin induced by IL-6 was found to lead to anemia associated with chronic inflammation [16]. A significant improvement at week 12 in hemoglobin levels from 11.3 g/dL to 12.8 g/dL in patients with RA patients treated with TCZ [87] was perhaps due to down-regulation of hepcidin, which was seen in patients with multicentric Castleman disease treated with TCZ [110].

Amyloid A amyloidosis is a serious complication of RA and amyloid fibril deposition causes progressive deterioration in various organs [17]. Since the activation of the SAA gene depends primarily on IL-6 [111,112], TCZ injection promptly reduces serum concentrations of SAA, just as in the case of CRP, and the suppressive effect of TCZ on serum levels of SAA appeared to be more powerful than that of TNF inhibitors [113]. Case reports and series studies to date have demonstrated the prominent ameliorative effect of TCZ on gastrointestinal symptoms [114-116], cardiac disease [117] or renal dysfunction [118] due to amyloid A amyloidosis complicated with RA, and amyloid A fibril deposits were found to have been eliminated in two cases after three injections of TCZ [114,116]. Moreover, a recent retrospective study demonstrated that TCZ was of greater clinical utility than TNF inhibitors in patients with amyloid A amyloidosis complicated with RA, evaluated by the ability to suppress SAA levels and the changes in the estimated glomerular filtration rate [119].

Cardiovascular events constitute the major determinant of poor prognosis for RA [20]. Lipid parameters such as high-density lipoprotein cholesterol, low-density lipoprotein cholesterol and triglyceride are often elevated during TCZ treatment but whether the increase is atherogenic remains to be determined [120]. In contrast, improvements due to TCZ treatment in serum biological markers related to atherosclerosis have been reported. IL-6 is reportedly involved in insulin resistance and development of type 2 diabetes mellitus [22,121,122], and we observed that TCZ treatment caused a reduction in HbA1c levels in diabetic patients with RA [123]. The average HbA1c level of diabetic patients (n=10) significantly decreased from 7.2% to 6.4% before steroid tapering and one month after tocilizumab treatment and to 6.0% after 6 months. Insulin resistance indices, such as the Homeostasis Model of Assessment-Insulin Resistance (HOMA-IR) index and the leptin-to-adiponectin ratio, also improved in non-diabetic patients with RA after a 3-month treatment with TCZ [124]. RA patients treated with TCZ showed significantly lower levels of reactive oxygen metabolites than those treated with DMARDs or TNF inhibitors (infliximab or etanecept) [125]. Moreover, endothelial dysfunction and aortic stiffness of RA patients were recently found to have improved as a result of TCZ treatment [126,127]. The causal relationship between IL-6R gene polymorphism and coronary heart disease [55,56] as mentioned earlier and the findings reported here indicate that long-term TCZ treatment may offer protection against cardiovascular events. A randomized, open-label, parallelgroup, multicenter study is in progress to evaluate the frequency of cardiovascular events following treatment with TCZ compared to that with etanercept for patients with RA.

These findings indicate the TCZ is efficacious for suppression of not only joint inflammation but also of systemic inflammation, thus leading to better prognosis.

Safety profile of tocilizumab

The safety profiles of TCZ monotherapy for Japanese RA patients were obtained from six initial trials and five long-term extensions [128]. For these studies, 601 patients with a total exposure to TCZ of 2,188 patient years (pt-yr) were enrolled. The median treatment duration was 3.8 years. The incidence of AEs, including abnormal laboratory test findings, was calculated as 465/100 pt-yr, with infections being the most common serious AEs (6.2/100 pt-yr). The results of an interim analysis of a post-marketing surveillance of all patients treated with TCZ in Japan were also reported [129]. This analysis comprised 3,881 patients who received 8 mg/kg of TCZ q4w and were observed for 28 weeks. Occurrence of a total of 3,004 AEs for 1,641 patients (167/100 ptyr) and 490 serious AEs for 361 patients (27/100 pt-yr) were recorded. The most frequent AE and serious AE was infection at 31/100 pt-yr and 9/100 pt-yr, respectively, with the majority of infections being pneumonia and cellulitis. Abnormalities in laboratory test findings, such as increases in lipid and liver function parameters were common and total and serious AEs associated with laboratory test abnormalities were 35/100 pt-yr and 2/100 pt-yr, respectively. While white blood cell and neutrophil counts usually decreased just after TCZ injection, this was not related to the incidence of infection. Twenty-five patients died for a standardized mortality ratio of 1.66, which was similar to the results reported for a Japanese cohort study of RA. The results of this analysis thus indicated that the safety profile of TCZ is acceptable for clinical settings.

The same post-marketing surveillance found seven cases of gastrointestinal (GI) perforation in six patients. In the worldwide Roche clinical trials, 26 (0.65%) cases of GI perforation were found among patients with RA treated with TCZ for a rate of 1.9/1,000 ptyr and most cases appeared to be complications of diverticulitis [130]. This rate is intermediate between the GI perforation rates of 3.9/1,000 pt-yr for corticosteroids and 1.3/1,000 pt-yr for anti-TNFα agents reported in the United Health Care database.

The reactivation of tuberculosis is a major concern during anti- TNF treatment [131], but there is no medical consensus regarding the effect of IL-6 blockade on tuberculosis. Okada et al. examined the effects of IL-6 and TNFα blockade on the development of tuberculosis infection in mice and observed that there was less tuberculosis infection for anti-IL-6R Ab than for anti-TNFα [132]. In addition, we were able to show that tuberculosis antigens-induced interferon (IFN)-γ production was suppressed by the addition of TNF inhibitors (infliximab and etanercept) but not of TCZ [133]. Although it seems likely that the incidence of reactivation of tuberculosis is lower during TCZ treatment than that during anti-TNF treatment, more detailed studies will be needed for further clarification.

Although dysregulated persistent production of IL-6 plays a pathological role in the development of RA, IL-6, when transiently produced in response to environmental stress such as infections and injuries, contributes to host defense [3-7,9,10]. Previously it was found that cutaneous wound healing in IL-6 gene-deficient mice was significantly slower than that in wild type control mice [134-136]. It was found that gene expression enhancement of IL-1, chemokines, adhesion molecules, TGF-β1 and VEGF at the wound sites was significantly reduced in IL-6-deficient mice [136]. Moreover, reduction of the wound area was delayed due to attenuated leukocyte infiltration, reepithelialization, angiogenesis and collagen accumulation. IL-6 is also important for tendon [137], bone fracture [138] and intestinal wound healing [139]. These findings indicate that TCZ treatment may result in delayed wound healing [140]. The TOPP study examined 161 Japanese orthopedic surgery cases during TCZ treatment [141] and found that infection rates of patients treated with TCZ were not noticeably high whereas incidence of delayed wound healing was indeed significantly higher in cases having undergone surgical interventions such as foot and spinal surgeries. Moreover, in patients treated with TCZ, the normal postoperative rise in temperature and CRP were both suppressed, thus potentially masking signs and symptoms of infection [142,143]. These observations suggest that, for reasons of safety, elective surgery needs planning for at least 2-3 weeks after the last administration of TCZ and a 2-week waiting period before restarting TCZ, since TCZ has an 11-13 day half-life depending on dose.

Mechanisms involved in the efficacy of tocilizumab

TCZ treatment led to improvements in serological or urinary markers related to bone and cartilage metabolisms [93,144,145]. The treatment also reduced circulating RANKL and VEGF concentrations [34,94] as well as RANKL expression in bone marrow tissues [145]. Moreover, several immunological analyses have been performed to clarify the mechanisms through which TCZ exerts its efficacy. Of particular importance is to determine whether TCZ can repair the Th17/Treg imbalance [13,146,147], which is thought to be a fundamental immunological abnormality in RA. It was recently shown that after three injections of TCZ in 15 patients with active RA, levels of Th17 (CD4+IL-17+) cells in peripheral blood decreased while Treg (CD4+CD25highFoxp3+) cells increased, indicating that inhibition of IL-6 function by TCZ corrected the imbalance between Th17 cells and Treg cells [148]. However, another study reported that in eight RA patients treated with TCZ for 24 weeks, the treatment did not induce any changes in the frequency of Th1 or Th17 cells in peripheral blood mononuclear cells but did increase the percentage of peripheral Treg cells [149]. Moreover, it was recently shown that tocilizumab caused a selective decrease of IL-21 production by memory/activated T cells in eight patients with RA as a result of 6-month treatment [150]. IL- 21 is known to promote plasma cell differentiation and induce IgG4 production while the tocilizumab treatment led to reduce serum levels of IgG4-specific ACPA, indicating the presence of a pathway involving IL-6, IL-21 and IgG4 autoantibodies in RA. Further evaluation will be required to clarify the effect of TCZ on the CD4 effector T cell subsets. In another study, Roll et al. examined 16 RA patients for the in vivo effect of TCZ on the B-cell compartment and found that it induced a significant reduction of peripheral pre-switch and post-switch memory B cells [151]. In addition, TCZ but not a TNF inhibitor (etanercept) significantly reduced somatic hypermutation in immunoglobulin gene rearrangements in pre-switch memory B cells [152], suggesting that modulation of memory B cells may be another possible target for TCZ.

Place of Tocilizumab in Biological Treatment of RA

A number of biologics are now available for the treatment of RA. These include TNF inhibitors (infliximab, etanercept, adalimumab, golimumab and certolizumab pegol), an IL-1 antagonist (anakinra), a B-cell depletor (rituximab) and a T-cell costimulation inhibitor (abatacept) as well as an IL-6R blocker (TCZ) [4]. It has not yet been determined which of these biologics should be selected for a given patient. Currently, one of the TNF inhibitors is recommended as a firstline biologic [153-155], but between 14 and 38% of patients show no or little response to anti-TNF treatment, with as many as 40% of patients discontinuing these drugs within a year and 50% within 2 years. As mentioned earlier, the RADIATE trial showed that RA patients who had previously discontinued TNF inhibitors achieved ACR20/50/70 responses of 50%, 28.8% and 12.4%, respectively, in response to TCZ [91]. In addition, the ACT-SURE [99], TAMARA [101,102], DANBIO [103] and REACTION [104,105] trials also showed that TCZ was efficacious for patients with RA who had been previously treated with TNF inhibitors. At present, TCZ is likely to be prescribed as a second-line biological therapy and will have to overcome significant competition from established anti-TNF therapies to be accepted as a first-line biologic. Meta-analyses of indirect comparisons of the efficacy of TCZ and other biologics have demonstrated that the efficacy of TCZ for patients with established RA is comparable to that of other biologics, including TNF inhibitors, abatacept and rituximab [155-159]. A comparison of AEs associated with these biologics in a review by Cochrane showed that the frequency of AEs, serious AEs and serious infections occurring during TCZ treatment was comparable to that for other biologics [160].

Biological treatment for RA patients is recommended to be initiated when patients show an inadequate response to MTX or to MTX combined with other DMARDs, and TNF inhibitors are currently positioned as the first biologic in combination with MTX [153,154]. On the other hand, patients who cannot tolerate sufficient doses of MTX could be treated with TCZ because TNF inhibitors need MTX to achieve their full efficacy [161-163], whereas the effect of TCZ monotherapy, as shown in the ACT-RAY trial, is not inferior to that of combination therapies using MTX [106]. Moreover, a clinical trial was recently performed involving a head-to-head comparison of biologics. The ADACTA study compared the monotherapeutic efficacy of TCZ with that of adalimumab [164]. 325 patients were randomly assigned to receive TCZ 8 mg/kg q4w intravenously plus placebo subcutaneously every 2 weeks (q2w) or adalimumab 40mg subcutaneously q2w plus placebo intravenously q4w for 24 weeks. Week 24 mean change from baseline in DAS28 was significantly greater in the TCZ group (-3.3) than in the adalimumab group (-1.8) patients (difference -1.5, 95% CI: -1.8 to -1.1; p<0.0001), indicating that TCZ as monotherapy is superior to adalimumab for reducing RA disease activity, although it remains to be determined whether TCZ combined with MTX is superior to adalimumab plus MTX. Further head-to-head comparative studies between TCZ and other biologics with or without MTX or other DMARDs are sure to determine the place of TCZ in the biological treatment of RA.

In addition, and of special importance, it can be expected that TCZ will become the first-line biologic for patients with serological positivity or systemic complications since IL-6 plays a central role in the fundamental immunological abnormality of RA and the development of systemic complications related to RA. However, further clinical studies and clarification of mechanisms through which TCZ exerts its clinical efficacy is essential to determine the optimal use of TCZ for RA patients.

Development of Subcutaneous Administration for Tocilizumab and Other IL-6 Inhibitors

TCZ is currently administered intravenously but TCZ also proved effective as a Subcutaneous (SC) administration [165-168]. The MATSURI study was a phase I/II study of SC infusion of TCZ (TCZSC) [165]. A total 32 patients were divided into three groups (81 mg q2w, 162 mg q2w and 162 mg weekly [qw]). Total treatment duration was 27 to 33 weeks. Neither serious AEs nor serious injection site reactions were observed. Cmax and area under the blood concentration time curve (AUC) after the first administration were 4.9 μg/mL and 444 μg hr/mL for 81 mg, and 10.9 μg/mL and 2,300 μg hr/mL for 162 mg. After 25 weeks, ACR20 response rates were 37.5%, 83.3% and 91.7% for 81 mg q2w, 162 mg q2w and 162 mg qw, respectively.

The MUSASHI study was a Japanese phase III study of TCZ-SC monotherapy [166]. Patients with RA who had responded inadequately to DMARDs or biologics except for TCZ were randomized to receive TCZ-SC 162 mg q2w or IV injection of TCZ (TCZ-IV) 8 mg/kg q4w without any DMARDs. The primary endpoint was to assess whether TCZ-SC was inferior or not to TCZ-IV in terms of ACR20 response rate at week 24 in per protocol patients. The non-inferiority margin was predefined at 18% (based on the SATORI study of monotherapy). A total 346 patients were enrolled (173 in TCZ-SC and 173 in TCZIV). Baseline demographics of the TCZ-SC and TCZ-IV groups were comparable, including body weight (53.8 ± 8.7 kg for TCZ-SC and 54.4 ± 10.1 kg for TCZ-IV). At week 24, 79.2% (95% CI: 72.9-85.5) of the TCZ-SC patients and 88.5% (95% CI: 83.4-93.5) of the TCZ-IV patients had attained an ACR20 response for a difference of -9.4% (95% CI: -17.6 to -1.2), thus confirming the non-inferiority of TCZ-SC to TCZIV. ACR50/70 response and DAS28/CDAI/Boolean remission rates were also similar for the two groups. Although safety profiles were also comparable, anti-TCZ Abs were detected more frequently in the TCZSC than in the TCZ-IV group. However, this anti-TCZ Abs did not affect the efficacy and were not related to anaphylactoid reaction.

The SUMMACTA study is a 2-year phase III study including a 24- week double-blind period, followed by a 72-week open-label phase. Patients were assigned to receive TCZ-SC 162 mg qw or TCZ-IV 8 mg/ kg q4w, in combination with traditional DMARDs [167]. The primary end point to determine whether TCZ-SC was inferior or not to TCZIV, was the percentage of patients in each group having met the ACR20 improvement criteria at week 24. The 1,262 patients enrolled in this study were stratified by body weight. Mean baseline characteristics of the TCZ-SC and TCZ-IV group were similar. The supposition of the non-inferiority of TCZ-SC was tested by means of 95% CI and with a non-inferiority margin (NIM) of 12%. At week 24, 69.4% (95% CI: 65.5-73.2) of the TCZ-SC-treated patients compared with 73.4% (95% CI: 69.6-77.1) of the TCZ-IV-treated patients had attained an ACR20 response (weighted difference between groups was 4.0% [95% CI: -9.2 to 1.2]), so that the 12% NIM was met. Disease activity, physical function improvements and safety profile of the two groups were also comparable.

The BREAVACTA is a 2-year; phase III, randomized, multicenter, parallel-group study [168]. A total of 656 patients were randomized to receive TCZ-SC 162 mg q2w or placebo-SC for 24 weeks, combined with stable doses of pre-study DMARDs. Significantly more patients receiving TCZ-SC had attained an ACR20 response at week 24, compared to those in the placebo-SC group (60.9% vs. 31.5%). Mean change in mTSS from baseline at week 24 was also significantly less for the former than the latter (0.62 ± 2.7 vs. 1.23 ± 2.8, p=0.0149). Injection site reactions occurred more frequently in the TCZ-SC group but hypersensitivity reactions were similar for the two groups. These studies demonstrated that subcutaneous administration of TCZ was efficacious for RA.

Furthermore, a new fully human Ab against IL-6R (SA237), generated from TCZ by means of Ab structural optimization technology, has been developed and this IgG2 class Ab significantly improved the pharmacokinetics and duration of CRP inhibition in cynomolgus monkeys [169].

The success of the clinical indication of TCZ for the treatment of RA has made it clear that targeting of IL-6 could be a new treatment strategy for RA, so that other IL-6 inhibitors are now being developed [170]. These include fully human anti-IL-6R Ab (REGN88/SAR153191 [sarilumab]), anti-IL-6R nanobody (ALX-0061), anti-IL-6 Ab (CNTO 136 [sirukumab], ALD518 [BMS-945429], CDP6038 [olokizumab], and MEDI5117) and soluble gp130-Fc fusion protein (FE301) (Table 1).

Agents Target Clinical trials
Sarilumab (REGN88/SAR153191) IL-6R Phase III
Sirukumab (CNTO 136) IL-6 Phase III
BMS-945429 (ALD518) IL-6 Phase II
Olokizumab (CDP6038) IL-6 Phase II
ALX-0061 IL-6R Phase II
MEDI5117 IL-6 Phase I
SA237 IL-6R Pre-clinical
FE301 (sgp130-Fc) Soluble IL-6R Pre-clinical

Table 1: New IL-6 targeting agents.

Sarilumab (REGN88/SAR153191) is a fully human monoclonal Ab directed against IL-6R. The phase II MOBILITY study, part A, involved 306 patients, who were randomized to a 12-week administration of sarilumab 100 mg qw, 150 mg qw, 100 mg q2w, 150 mg q2w, 200 mg q2w or placebo added to stable MTX [171]. An ACR20 response after 12 weeks was seen in 49.0% of the patients receiving the lowest sarilumab dose regime and in 72.0% of the patients receiving the highest dose regime compared to 46.2% of those treated with placebo and MTX. The types and incidence of AEs were consistent with those previously reported for TCZ. The phase III MOBILITY study, part B, for RA (SARIL-RA-MOBILITY) is ongoing.

ALX-0061 is a 26kD bi-specific IL-6R targeting nanobody with monovalent binding to IL-6R and serum albumin. Nanobodies are therapeutic proteins based on the smallest functional fragments of heavy chain antibodies, naturally occurring in Camelidae, which possess a high degree of homology to human Ig VH domains. In a phase I/II trial of ALX-0061, 28 patients with active RA were included [172]. Patients were assigned to receive ALX-0061 3 mg/kg q4w, 6 mg/kg q8w or placebo added to stable MTX therapy for 24 weeks. ACR20/50/70 response rates were 80%/50%/10% for the 3 mg/kg q4w group, 56%/56%/44% for the group receiving 6 mg/kg q8w of ALX- 0061 and 17%/0%/0% for the placebo-injected group.

Sirukumab (CNTO 136) is a human monoclonal Ab with high affinity and specificity for binding to IL-6. After completion of a phase I trial [173,174], 151 patients were enrolled in a phase II study [175]. The patients were randomized equally to receive SC injections of placebo q2w for weeks 0-10 and sirukumab 100 mg 2qw for weeks 12- 24, and sirukumab 100 mg q2w, 100 mg q4w, 50 mg q4w or 25 mg q4w for weeks 0-24. At week 12 (pre-crossover), more patients receiving sirukumab were in remission than those given the placebo according to both Boolean- and SDAI-based ACR/EULAR criteria (2% vs. 0% and 6% vs. 3%). At week 24, high remission rates determined with ACR/ EULAR or DAS28 (CRP) criteria had been attained with sirukumab at SC dose regimens ranging from 25-100 mg q2w-q4w. The types and incidence of AEs were consistent with those previously reported for TCZ [176]. A phase III trial of sirukumab for RA (the SIRROUND study) is ongoing.

BMS-945429 (ALD518) is an asialated, humanized anti-IL-6 monoclonal Ab with a prolonged half-life and assessed in a phase II study with 127 patients enrolled. Patients were randomized to receive IV injections of ALD518 80 mg q8w, 160 mg q8w, 320 mg q8w or placebo for 16 weeks. ACR20/50/70 response rates at 16 weeks were 75%/41%/22% for the 80 mg q8w, 65%/41%/18% for the 160 mg q8w, 82%/50%/43% for the 320 mg q8w and 36%/15%/6% for the placebo group, respectively [177]. A phase II study of SC injection of ALD518 (200 mg q4w or 100 mg q4w with or without MTX) is now in progress.

Olokizumab (CDP6038) is a humanized monoclonal anti-IL-6 Ab. A phase I study demonstrated that olokizumab was tolerated at single doses of up to 3 mg/kg SC and 10 mg/kg IV with a median half-life of 31.1 days [178]. In a phase II trial to evaluate the efficacy and safety of olokizumab administered SC for 12 weeks, 220 patients were enrolled in this randomized, double-blind, placebo-controlled, dose-ranging study with TCZ as an active comparator. Patients were randomized to receive olokizumab 60 mg q2w, 60 mg q4w, 120 mg q2w, 120 mg q4w, 240 mg q4w or placebo. All patients received concomitant MTX treatment. A significant reduction in DAS28 at week 12 was observed for all olokizumab-treated groups relative to the placebo group. The exploratory analyses of these data suggest that the efficacy and safety of olokizumab and TCZ are comparable. A trial to determine the longterm tolerability of 120 mg SC olokizumab is also ongoing.

Another human anti-IL-6 Ab, MEDI5117, was developed in China. This IL-6 inhibitor was generated by means of variable domain engineering to achieve sub-picomolar affinity for IL-6 and with Fc engineering to enhance its pharmacokinetic half-life [179]. A phase I trial of IV injection of MEDI5117 (30 mg, 100 mg, 300 mg or 600 mg) for RA is in progress.

FE301, a fusion protein consisting of the extracellular region of gp130 and the Fc part of a human IgG1 Ab (sgp130-Fc) selectively inhibits IL-6 trans-signaling without affecting responses via the membrane-bound IL-6R [180]. Trans-signaling of IL-6 (signaling via soluble IL-6R) mainly activates various inflammatory cells, which do not express transmembrane IL-6R but express gp130, whereas classicsignaling via transmembrane IL-6R, which is expressed on cells limited to monocytes, macrophages and hepatocytes, mainly activates immune response [170]. FE301 can therefore be expected to prove to be an antiinflammatory agent, which does not affect immunosuppression nor interferes with acute phase response. The clinical utility of FE301 is as yet unknown.

Widespread Use of Tocilizumab for Various Immune-Mediated Diseases

TCZ is now used as an innovative drug for RA in more than 100 countries worldwide and in several countries for systemic juvenile idiopathic arthritis, polyarticular juvenile idiopathic arthritis and Castleman’s disease [4-7,181]. Because of the pathological role of IL-6 in various immune-mediated diseases, TCZ is expected to be used widely for the treatment of these and other diseases and favorable results of recent clinical trials and case reports have suggested that this is indeed a realistic expectation [146,147,182]. These other diseases include systemic autoimmune diseases such as systemic lupus erythematosus, systemic sclerosis, polymyositis, vasculitis syndrome, relapsing polychondritis and Cogan syndrome in addition to organspecific autoimmune diseases including autoimmune hemolytic anemia, acquired hemophilia A, and neuromyelitis optica, as well as chronic inflammatory diseases such as adult-onset Still’s disease, Crohn’s disease, polymyalgia rheumatica, remitting seronegative, symmetrical synovitis with pitting edema, graft-versus-host disease, Behcet’s disease, uveitis, Schnitzler syndrome, TNF-associated periodic syndrome, chronic infantile neurological cutaneous and articular syndrome, pulmonary arterial hypertension, as well as sciatica and atopic dermatitis. Some case studies have reported that TCZ was efficacious for spondyloarthritides such as ankylosing spondylitis and reactive arthritis, although recent clinical trials of TCZ as well as of sarilumab could not detect any beneficial effects for ankylosing spondylitis [183,184]. Further clinical trials to evaluate the efficacy and safety of TCZ for its widespread use for the treatment of various diseases will therefore be essential.

Conclusion

Many clinical studies have proved that IL-6 blockade treatment is efficacious for the treatment of RA and TCZ, a humanized anti-IL-6R Ab, is being used worldwide as an innovative drug for RA patients. Since several other biological agents have also been approved, the place of TCZ in biological treatment needs to be clarified. Present evidence suggests that TCZ can be recommended for RA patients who cannot tolerate sufficient doses of MTX, but further clinical studies are needed to clarify the best use of TCZ. Moreover, in view of the pleiotropic activity of IL-6, the strategy of targeting IL-6 can be expected to be applicable to the treatment of a wide variety of immune-mediated diseases and a number of clinical trials to verify this potential are in progress.

Acknowledgements

T. Tanaka has received grant and payment for lectures including service on speaker’s bureaus from Chugai Pharmaceutical Co, Ltd. A. Ogata has received a consulting fee as a medical adviser, grant, and payment for lectures including service on speaker’s bureaus from Chugai Pharmaceutical Co, Ltd. T. Kishimoto holds a patent for tocilizumab and has received royalties for Actemra.

References

  1. Smolen JS, Aletaha D, Koeller M, Weisman MH, Emery P (2007) New therapies for treatment of rheumatoid arthritis. Lancet 370: 1861-1874.
  2. McInnes IB, Schett G (2011) The pathogenesis of rheumatoid arthritis. N Engl J Med 365: 2205-2219.
  3. Kishimoto T (2005) Interleukin-6: from basic science to medicine--40 years in immunology. Annu Rev Immunol 23: 1-21.
  4. Tanaka T, Ogata A, Narazaki M (2010) Tocilizumab for the treatment of rheumatoid arthritis. Expert Rev Clin Immunol 6: 843-854.
  5. Tanaka T, Kishimoto T (2011) Immunotherapy of tocilizumab for rheumatoid arthritis. J Clin Cell Immunol S6-001.
  6. Hashizume M, Mihara M (2011) The roles of interleukin-6 in the pathogenesis of rheumatoid arthritis. Arthritis 2011: 765624.
  7. Ogata A, Tanaka T (2012) Tocilizumab for the treatment of rheumatoid arthritis and other systemic autoimmune diseases: current perspectives and future directions. Int J Rheumatol 2012: 946048.
  8. Hirano T, Yasukawa K, Harada H, Taga T, Watanabe Y, et al. (1986) Complementary DNA for a novel human interleukin (BSF-2) that induces B lymphocytes to produce immunoglobulin. Nature 324: 73-76.
  9. Akira S, Taga T, Kishimoto T (1993) Interleukin-6 in biology and medicine. Adv Immunol 54: 1-78.
  10. Suematsu S, Matsuda T, Aozasa K, Akira S, Nakano N, et al. (1989) IgG1 plasmacytosis in interleukin 6 transgenic mice. Proc Natl Acad Sci U S A 86: 7547-7551.
  11. Korn T, Bettelli E, Oukka M, Kuchroo VK (2009) IL-17 and Th17 Cells. Annu Rev Immunol 27: 485-517.
  12. Kimura A, Kishimoto T (2010) IL-6: regulator of Treg/Th17 balance. Eur J Immunol 40: 1830-1835.
  13. Heinrich PC, Castell JV, Andus T (1990) Interleukin-6 and the acute phase response. Biochem J 265: 621-636.
  14. Siewert E, Bort R, Kluge R, Heinrich PC, Castell J, et al. (2000) Hepatic cytochrome P450 down-regulation during aseptic inflammation in the mouse is interleukin 6 dependent. Hepatology 32: 49-55.
  15. Nemeth E, Rivera S, Gabayan V, Keller C, Taudorf S, et al. (2004) IL-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin. J Clin Invest 113: 1271-1276.
  16. Obici L, Merlini G (2012) AA amyloidosis: basic knowledge, unmet needs and future treatments. Swiss Med Wkly 142: w13580.
  17. Ishibashi T, Kimura H, Shikama Y, Uchida T, Kariyone S, et al. (1989) Interleukin-6 is a potent thrombopoietic factor in vivo in mice. Blood 74: 1241-1244.
  18. Kim S, Östör AJ, Nisar MK (2012) Interleukin-6 and cytochrome-P450, reason for concern? Rheumatol Int 32: 2601-2604.
  19. Meune C, Touzé E, Trinquart L, Allanore Y (2009) Trends in cardiovascular mortality in patients with rheumatoid arthritis over 50 years: a systematic review and meta-analysis of cohort studies. Rheumatology (Oxford) 48: 1309-1313.
  20. Abeywardena MY, Leifert WR, Warnes KE, Varghese JN, Head RJ (2009) Cardiovascular biology of interleukin-6. Curr Pharm Des 15: 1809-1821.
  21. Schuett H, Luchtefeld M, Grothusen C, Grote K, Schieffer B (2009) How much is too much? Interleukin-6 and its signalling in atherosclerosis. Thromb Haemost 102: 215-222.
  22. Rohleder N, Aringer M, Boentert M (2012) Role of interleukin-6 in stress, sleep, and fatigue. Ann N Y Acad Sci 1261: 88-96.
  23. Schaap LA, Pluijm SM, Deeg DJ, Visser M (2006) Inflammatory markers and loss of muscle mass (sarcopenia) and strength. Am J Med 119: 526.
  24. Pereira LS, Narciso FM, Oliveira DM, Coelho FM, Souza Dda G, et al. (2009) Correlation between manual muscle strength and interleukin-6 (IL-6) plasma levels in elderly community-dwelling women. Arch Gerontol Geriatr 48: 313-316.
  25. Romano M, Sironi M, Toniatti C, Polentarutti N, Fruscella P, et al. (1997) Role of IL-6 and its soluble receptor in induction of chemokines and leukocyte recruitment. Immunity 6: 315-325.
  26. Suzuki M, Hashizume M, Yoshida H, Mihara M (2010) Anti-inflammatory mechanism of tocilizumab, a humanized anti-IL-6R antibody: effect on the expression of chemokine and adhesion molecule. Rheumatol Int 30: 309-315.
  27. Guerne PA, Zuraw BL, Vaughan JH, Carson DA, Lotz M (1989) Synovium as a source of interleukin 6 in vitro. Contribution to local and systemic manifestations of arthritis. J Clin Invest 83: 585-592.
  28. Mihara M, Moriya Y, Kishimoto T, Ohsugi Y (1995) Interleukin-6 (IL-6) induces the proliferation of synovial fibroblastic cells in the presence of soluble IL-6 receptor. Br J Rheumatol 34: 321-325.
  29. Kotake S, Sato K, Kim KJ, Takahashi N, Udagawa N, et al. (1996) Interleukin-6 and soluble interleukin-6 receptors in the synovial fluids from rheumatoid arthritis patients are responsible for osteoclast-like cell formation. J Bone Miner Res 11: 88-95.
  30. Hashizume M, Hayakawa N, Mihara M (2008) IL-6 trans-signalling directly induces RANKL on fibroblast-like synovial cells and is involved in RANKL induction by TNF-alpha and IL-17. Rheumatology (Oxford) 47: 1635-1640.
  31. Poli V, Balena R, Fattori E, Markatos A, Yamamoto M, et al. (1994) Interleukin-6 deficient mice are protected from bone loss caused by estrogen depletion. EMBO J 13: 1189-1196.
  32. Edwards CJ, Williams E (2010) The role of interleukin-6 in rheumatoid arthritis-associated osteoporosis. Osteoporos Int 21: 1287-1293.
  33. Nakahara H, Song J, Sugimoto M, Hagihara K, Kishimoto T, et al. (2003) Anti-interleukin-6 receptor antibody therapy reduces vascular endothelial growth factor production in rheumatoid arthritis. Arthritis Rheum 48: 1521-1529.
  34. Hashizume M, Hayakawa N, Suzuki M, Mihara M (2009) IL-6/sIL-6R trans-signalling, but not TNF-alpha induced angiogenesis in a HUVEC and synovial cell co-culture system. Rheumatol Int 29: 1449-1454.
  35. Silacci P, Dayer JM, Desgeorges A, Peter R, Manueddu C, et al. (1998) Interleukin (IL)-6 and its soluble receptor induce TIMP-1 expression in synoviocytes and chondrocytes, and block IL-1-induced collagenolytic activity. J Biol Chem 273: 13625-13629.
  36. Suzuki M, Hashizume M, Yoshida H, Shiina M, Mihara M (2010) IL-6 and IL-1 synergistically enhanced the production of MMPs from synovial cells by up-regulating IL-6 production and IL-1 receptor I expression. Cytokine 51: 178-183.
  37. Hirano T, Matsuda T, Turner M, Miyasaka N, Buchan G, et al. (1988) Excessive production of interleukin 6/B cell stimulatory factor-2 in rheumatoid arthritis. Eur J Immunol 18: 1797-1801.
  38. Houssiau FA, Devogelaer JP, Van Damme J, de Deuxchaisnes CN, Van Snick J (1988) Interleukin-6 in synovial fluid and serum of patients with rheumatoid arthritis and other inflammatory arthritides. Arthritis Rheum 31: 784-788.
  39. Robak T, Gladalska A, StepieÅ, H, Robak E (1998) Serum levels of interleukin-6 type cytokines and soluble interleukin-6 receptor in patients with rheumatoid arthritis. Mediators Inflamm 7: 347-353.
  40. Holt I, Cooper RG, Hopkins SJ (1991) Relationships between local inflammation, interleukin-6 concentration and the acute phase protein response in arthritis patients. Eur J Clin Invest 21: 479-484.
  41. Dasgupta B, Corkill M, Kirkham B, Gibson T, Panayi G (1992) Serial estimation of interleukin 6 as a measure of systemic disease in rheumatoid arthritis. J Rheumatol 19: 22-25.
  42. Madhok R, Crilly A, Watson J, Capell HA (1993) Serum interleukin 6 levels in rheumatoid arthritis: correlations with clinical and laboratory indices of disease activity. Ann Rheum Dis 52: 232-234.
  43. Straub RH, Müller-Ladner U, Lichtinger T, Schölmerich J, Menninger H, et al. (1997) Decrease of interleukin 6 during the first 12 months is a prognostic marker for clinical outcome during 36 months treatment with disease-modifying anti-rheumatic drugs. Br J Rheumatol 36: 1298-1303.
  44. Fishman D, Faulds G, Jeffery R, Mohamed-Ali V, Yudkin JS, et al. (1998) The effect of novel polymorphisms in the interleukin-6 (IL-6) gene on IL-6 transcription and plasma IL-6 levels, and an association with systemic-onset juvenile chronic arthritis. J Clin Invest 102: 1369-1376.
  45. Pascual M, Nieto A, Matarán L, Balsa A, Pascual-Salcedo D, et al. (2000) IL-6 promoter polymorphisms in rheumatoid arthritis. Genes Immun 1: 338-340.
  46. Lee YH, Bae SC, Choi SJ, Ji JD, Song GG (2012) The association between interleukin-6 polymorphisms and rheumatoid arthritis: a meta-analysis. Inflamm Res 61: 665-671.
  47. Marinou I, Healy J, Mewar D, Moore DJ, Dickson MC, et al. (2007) Association of interleukin-6 and interleukin-10 genotypes with radiographic damage in rheumatoid arthritis is dependent on autoantibody status. Arthritis Rheum 56: 2549-2556.
  48.  Pawlik A, Wrzesniewska J, Florczak M, Gawronska-Szklarz B, Herczynska M (2005) IL-6 promoter polymorphism in patients with rheumatoid arthritis. Scand J Rheumatol 34: 109-113.
  49. Kim LH, Lee HS, Kim YJ, Jung JH, Kim JY, et al. (2003) Identification of novel SNPs in the interleukin 6 receptor gene (IL6R). Hum Mutat 21: 450-451.
  50. Galicia JC, Tai H, Komatsu Y, Shimada Y, Akazawa K, et al. (2004) Polymorphisms in the IL-6 receptor (IL-6R) gene: strong evidence that serum levels of soluble IL-6R are genetically influenced. Genes Immun 5: 513-516.
  51. Rafiq S, Frayling TM, Murray A, Hurst A, Stevens K, et al. (2007) A common variant of the interleukin 6 receptor (IL-6r) gene increases IL-6r and IL-6 levels, without other inflammatory effects. Genes Immun 8: 552-559.
  52. Rodríguez-Rodríguez L, Lamas JR, Varadé J, López-Romero P, Tornero-Esteban P, et al. (2011) Plasma soluble IL-6 receptor concentration in rheumatoid arthritis: associations with the rs8192284 IL6R polymorphism and with disease activity. Rheumatol Int 31: 409-413.
  53. Marinou I, Walters K, Winfield J, Bax DE, Wilson AG (2010) A gain of function polymorphism in the interleukin 6 receptor influences RA susceptibility. Ann Rheum Dis 69: 1191-1194.
  54. IL6R Genetics Consortium Emerging Risk Factors Collaboration, Sarwar N, Butterworth AS, Freitag DF, Gregson J, et al. (2012) Interleukin-6 receptor pathways in coronary heart disease: a collaborative meta-analysis of 82 studies. Lancet 379: 1205-1213.
  55. Interleukin-6 Receptor Mendelian Randomisation Analysis (IL6R MR) Consortium, Hingorani AD, Casas JP (2012) The interleukin-6 receptor as a target for prevention of coronary heart disease: a mendelian randomisation analysis. Lancet 379: 1214-1224.
  56. Boekholdt SM, Stroes ES (2012) The interleukin-6 pathway and atherosclerosis. Lancet 379: 1176-1178.
  57. Scheller J, Rose-John S (2012) The interleukin 6 pathway and atherosclerosis. Lancet 380: 338.
  58. Alonzi T, Fattori E, Lazzaro D, Costa P, Probert L, et al. (1998) Interleukin 6 is required for the development of collagen-induced arthritis. J Exp Med 187: 461-468.
  59. Takagi N, Mihara M, Moriya Y, Nishimoto N, Yoshizaki K, et al. (1998) Blockage of interleukin-6 receptor ameliorates joint disease in murine collagen-induced arthritis. Arthritis Rheum 41: 2117-2121.
  60. Sasai M, Saeki Y, Ohshima S, Nishioka K, Mima T, et al. (1999) Delayed onset and reduced severity of collagen-induced arthritis in interleukin-6-deficient mice. Arthritis Rheum 42: 1635-1643.
  61. Fujimoto M, Serada S, Mihara M, Uchiyama Y, Yoshida H, et al. (2008) Interleukin-6 blockade suppresses autoimmune arthritis in mice by the inhibition of inflammatory Th17 responses. Arthritis Rheum 58: 3710-3719.
  62. Ohshima S, Saeki Y, Mima T, Sasai M, Nishioka K, et al. (1998) Interleukin 6 plays a key role in the development of antigen-induced arthritis. Proc Natl Acad Sci U S A 95: 8222-8226.
  63. Wong PK, Quinn JM, Sims NA, van Nieuwenhuijze A, Campbell IK, et al. (2006) Interleukin-6 modulates production of T lymphocyte-derived cytokines in antigen-induced arthritis and drives inflammation-induced osteoclastogenesis. Arthritis Rheum 54: 158-168.
  64. Sakaguchi N, Takahashi T, Hata H, Nomura T, Tagami T, et al. (2003) Altered thymic T-cell selection due to a mutation of the ZAP-70 gene causes autoimmune arthritis in mice. Nature 426: 454-460.
  65. Hata H, Sakaguchi N, Yoshitomi H, Iwakura Y, Sekikawa K, et al. (2004) Distinct contribution of IL-6, TNF-alpha, IL-1, and IL-10 to T cell-mediated spontaneous autoimmune arthritis in mice. J Clin Invest 114: 582-588.
  66. Hirota K, Hashimoto M, Yoshitomi H, Tanaka S, Nomura T, et al. (2007) T cell self-reactivity forms a cytokine milieu for spontaneous development of IL-17+ Th cells that cause autoimmune arthritis. J Exp Med 204: 41-47.
  67. Iwakura Y, Tosu M, Yoshida E, Takiguchi M, Sato K, et al. (1991) Induction of inflammatory arthropathy resembling rheumatoid arthritis in mice transgenic for HTLV-I. Science 253: 1026-1028.
  68.  Iwakura Y, Saijo S, Kioka Y, Nakayama-Yamada J, Itagaki K, et al. (1995) Autoimmunity induction by human T cell leukemia virus type 1 in transgenic mice that develop chronic inflammatory arthropathy resembling rheumatoid arthritis in humans. J Immunol 155: 1588-1598.
  69. Atsumi T, Ishihara K, Kamimura D, Ikushima H, Ohtani T, et al. (2002) A point mutation of Tyr-759 in interleukin 6 family cytokine receptor subunit gp130 causes autoimmune arthritis. J Exp Med 196: 979-990.
  70. Sawa S, Kamimura D, Jin GH, Morikawa H, Kamon H, et al. (2006) Autoimmune arthritis associated with mutated interleukin (IL)-6 receptor gp130 is driven by STAT3/IL-7-dependent homeostatic proliferation of CD4+ T cells. J Exp Med 203: 1459-1470.
  71. Ogura H, Murakami M, Okuyama Y, Tsuruoka M, Kitabayashi C, et al. (2008) Interleukin-17 promotes autoimmunity by triggering a positive-feedback loop via interleukin-6 induction. Immunity 29: 628-636.
  72. Murakami M, Okuyama Y, Ogura H, Asano S, Arima Y, et al. (2011) Local microbleeding facilitates IL-6- and IL-17-dependent arthritis in the absence of tissue antigen recognition by activated T cells. J Exp Med 208: 103-114.
  73. Kagari T, Doi H, Shimozato T (2002) The importance of IL-1 beta and TNF-alpha, and the noninvolvement of IL-6, in the development of monoclonal antibody-induced arthritis. J Immunol 169: 1459-1466.
  74. Iwanami K, Matsumoto I, Tanaka-Watanabe Y, Inoue A, Mihara M, et al. (2008) Crucial role of the interleukin-6/interleukin-17 cytokine axis in the induction of arthritis by glucose-6-phosphate isomerase. Arthritis Rheum 58: 754-763.
  75. Kimura A, Naka T, Nohara K, Fujii-Kuriyama Y, Kishimoto T (2008) Aryl hydrocarbon receptor regulates Stat1 activation and participates in the development of Th17 cells. Proc Natl Acad Sci U S A 105: 9721-9726.
  76. Puga A, Tomlinson CR, Xia Y (2005) Ah receptor signals cross-talk with multiple developmental pathways. Biochem Pharmacol 69: 199-207.
  77. Nakahama T, Kimura A, Nguyen NT, Chinen I, Hanieh H, et al. (2011) Aryl hydrocarbon receptor deficiency in T cells suppresses the development of collagen-induced arthritis. Proc Natl Acad Sci U S A 108: 14222-14227.
  78. Yamasaki K, Taga T, Hirata Y, Yawata H, Kawanishi Y, et al. (1988) Cloning and expression of the human interleukin-6 (BSF-2/IFN beta 2) receptor. Science 241: 825-828.
  79. Hibi M, Murakami M, Saito M, Hirano T, Taga T, et al. (1990) Molecular cloning and expression of an IL-6 signal transducer, gp130. Cell 63: 1149-1157.
  80. Taga T, Kishimoto T (1997) Gp130 and the interleukin-6 family of cytokines. Annu Rev Immunol 15: 797-819.
  81. Murakami M, Hibi M, Nakagawa N, Nakagawa T, Yasukawa K, et al. (1993) IL-6-induced homodimerization of gp130 and associated activation of a tyrosine kinase. Science 260: 1808-1810.
  82. Boulanger MJ, Chow DC, Brevnova EE, Garcia KC (2003) Hexameric structure and assembly of the interleukin-6/IL-6 alpha-receptor/gp130 complex. Science 300: 2101-2104.
  83. Sato K, Tsuchiya M, Saldanha J, Koishihara Y, Ohsugi Y, et al. (1993) Reshaping a human antibody to inhibit the interleukin 6-dependent tumor cell growth. Cancer Res 53: 851-856.
  84. Choy EH, Isenberg DA, Garrood T, Farrow S, Ioannou Y, et al. (2002) Therapeutic benefit of blocking interleukin-6 activity with an antiinterleukin-6 receptor monoclonal antibody in rheumatoid arthritis: a randomized, double-blind, placebo-controlled, dose-escalation trial. Arthritis Rheum 46: 143-150.
  85. Nishimoto N, Yoshizaki K, Maeda K, Kuritani T, Deguchi H, et al. (2003) Toxicity, pharmacokinetics, and dose-finding study of repetitive treatment with the humanized anti-interleukin 6 receptor antibody MRA in rheumatoid arthritis. Phase I/II clinical study. J Rheumatol 30: 1426-1435.
  86. Nishimoto N, Yoshizaki K, Miyasaka N, Yamamoto K, Kawai S, et al. (2004) Treatment of rheumatoid arthritis with humanized anti-interleukin-6 receptor antibody: a multicenter, double-blind, placebo-controlled trial. Arthritis Rheum 50: 1761-1769.
  87. Maini RN, Taylor PC, Szechinski J, Pavelka K, Broll J, et al; CHARISMA Study Group (2006) 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 Rheum 54: 2817-2829.
  88. Nishimoto N, Hashimoto J, Miyasaka N, Yamamoto K, Kawai S, et al. (2007) 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. Ann Rheum Dis 66: 1162-1167.
  89. Genovese MC, McKay JD, Nasonov EL, Mysler EF, da Silva NA, et al. (2008) 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 58: 2968-2980.
  90. Emery P, Keystone E, Tony HP, Cantagrei A, van Vollenhoven R, et al. (2008) IL-6 receptor inhibition with tocilizumab improves treatment outcomes in patients with rheumatoid arthritis refractory to anti-tumor necrosis factor biologicals: results from a 24-week multicentre randomized placebo controlled trial. Ann Rheum Dis 67: 1516-1523.
  91. Smolen JS, Beaulieu A, Rubbert-Roth A, Ramos-Remus C, Rovensky J, et al; OPTION Investigators. (2008) Effect of interleukin-6 receptor inhibition with tocilizumab in patients with rheumatoid arthritis (OPTION study): a double-blind, placebo-controlled, randomized trial. Lancet 371: 987-997.
  92.  Garnero P, Thompson E, Woodworth T, Smolen JS. (2010) Rapid and sustained improvement in bone and cartilage turnover markers with the anti-interleukin-6 receptor inhibitor tocilizumab plus methotrexate in rheumatoid arthritis patients with an inadequate response to methotrexate: results from a substudy of the multicenter double-blind, placebo-controlled trial of tocilizumab in inadequate responders to methotrexate alone. Arthritis Rheum 62: 33-43.
  93. Nishimoto N, Miyasaka N, Yamamoto K, Kawai, S, Takeuchi T, et al. (2009) Study of active controlled tocilizumab monotherapy for rheumatoid arthritis patients with an inadequate response to methotrexate (SATORI): significant reduction in disease activity and serum vascular endothelial growth factor by IL-6 receptor inhibition therapy. Mod Rheumatol 19: 12-19.
  94.  Kremer JM, Blanco R, Brzosko M, Burgos-Vargas R, Halland AM, et al. (2011) 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 63: 609-621.
  95. Fleischmann RM, Halland AM, Brzosko M, Burgos-Vargas R, Mela C, et al. (2013) Tocilizumab inhibits structural joint damage and improves physical function in patients with rheumatoid arthritis and inadequate responses to methotrexate: LITHE study 2-year results. J Rheumatol 40: 113-126.
  96. Jones G, Sebba A, Gu J, Lowenstein MB, Calvo A, et al. (2010) Comparison of tocilizumab monotherapy versus methotrexate monotherapy in patients with moderate to severe rheumatoid arthritis: the AMBITION study. Ann Rheum Dis 69: 88-96.
  97. Yazici Y, Curtis JR, Ince A, Baraf H, Malamet RL, et al. (2012) Efficacy of tocilizumab in patients with moderate to severe active rheumatoid arthritis and a previous inadequate response to disease-modifying antirheumatic drugs: the ROSE study. Ann Rheum Dis 71: 198-205.
  98. Bykerk VP, Ostör AJ, Alvaro-Gracia J, Pavelka K, Ivorra JA, et al. (2012) Tocilizumab in patients with active rheumatoid arthritis and inadequate responses to DMARDs and/or TNF inhibitors: a large, open-label study close to clinical practice. Ann Rheum Dis 71: 1950-1954.
  99. Weinblatt ME, Kremer J, Cush J, Rigby W, Teng LL, et al. (2013) Tocilizumab as monotherapy or in combination with nonbiologic disease-modifying antirheumatic drugs: twenty-four-week results of an open-label, clinical practice study. Arthritis Care Res (Hoboken) 65: 362-371.
  100. Burmester GR, Feist E, Kellner H, Braun J, Iking-Konert C, et al. (2011) Effectiveness and safety of the interleukin 6-receptor antagonist tocilizumab after 4 and 24 weeks in patients with active rheumatoid arthritis: the first phase IIIb real-life study (TAMARA). Ann Rheum Dis 70: 755-759.
  101. Iking-Konert C, Aringer M, Wollenhaupt J, Mosch T, Tuerk S, et al. (2011) Performance of the new 2011 ACR/EULAR remission criteria with tocilizumab using the phase IIIb study TAMARA as an example and their comparison with traditional remission criteria. Ann Rheum Dis 70: 1986-1990.
  102. Leffers HC, Ostergaard M, Glintborg B, Krogh NS, Foged H, et al. (2011) Efficacy of abatacept and tocilizumab in patients with rheumatoid arthritis treated in clinical practice: results from the nationwide Danish DANBIO registry. Ann Rheum Dis 70: 1216-1222.
  103. Yamanaka H, Tanaka Y, Inoue E, Hoshi D, Momohara S, et al. (2011) Efficacy and tolerability of tocilizumab in rheumatoid arthritis patients seen in daily clinical practice in Japan: results from a retrospective study (REACTION study). Mod Rheumatol 21: 122-133.
  104. Takeuchi T, Tanaka Y, Amano K, Hoshi D, Nawata M, et al. (2011) Clinical, radiographic and functional effectiveness of tocilizumab for rheumatoid arthritis patients--REACTION 52-week study. Rheumatology (Oxford) 50: 1908-1915.
  105. Dougados M, Kissel K, Sheeran T, Tak PP, Conaghan PG, et al. (2013) Adding tocilizumab or switching to tocilizumab monotherapy in methotrexate inadequate responders: 24-week symptomatic and structural results of a 2-year randomised controlled strategy trial in rheumatoid arthritis (ACT-RAY). Ann Rheum Dis 72: 43-50.
  106. Strand V, Burmester GR, Ogale S, Devenport J, John A, et al. (2012) Improvements in health-related quality of life after treatment with tocilizumab in patients with rheumatoid arthritis refractory to tumour necrosis factor inhibitors: results from the 24-week randomized controlled RADIATE study. Rheumatology (Oxford) 51: 1860-1869.
  107. Fusama M, Nakahara H, Hamano Y, Nishide M, Kawamoto K, et al. (2013) Improvement of health status evaluated by Arthritis Impact Measurement Scale 2 (AIMS-2) and Short Form-36 (SF-36) in patients with rheumatoid arthritis treated with tocilizumab. Mod Rheumatol 23: 276-283.
  108. Fragiadaki K, Tektonidou MG, Konsta M, Chrousos GP, Sfikakis PP (2012) Sleep disturbances and interleukin 6 receptor inhibition in rheumatoid arthritis. J Rheumatol 39: 60-62.
  109. Song SN, Tomosugi N, Kawabata H, Ishikawa T, Nishikawa T, et al. (2010) Down-regulation of hepcidin resulting from long-term treatment with an anti-IL-6 receptor antibody (tocilizumab) improves anemia of inflammation in multicentric Castleman disease. Blood 116: 3627-3634.
  110. Hagihara K, Nishikawa T, Isobe T, Song J, Sugamata Y, et al. (2004) IL-6 plays a critical role in the synergistic induction of human serum amyloid A (SAA) gene when stimulated with proinflammatory cytokines as analyzed with an SAA isoform real-time quantitative RT-PCR assay system. Biochem Biophys Res Commun 314: 363-369.
  111. Hagihara K, Nishikawa T, Sugamata Y, Song J, Isobe T, et al. (2005) Essential role of STAT3 in cytokine-driven NF-kappaB-mediated serum amyloid A gene expression. Genes Cells 10: 1051-1063.
  112. Tanaka T, Hagihara K, Hishitani Y, Ogata A (2011) Tocilizumab for the treatment of AA amyloidosis: Amyloidosis - An insight to disease of systems and novel therapies edited by Isil Adadan Guvenc. INTECH Open Access Publisher. Croatia.
  113. Nishida S, Hagihara K, Shima Y, Kawai M, Kuwahara Y, et al. (2009) Rapid improvement of AA amyloidosis with humanised anti-interleukin 6 receptor antibody treatment. Ann Rheum Dis 68: 1235-1236.
  114. Sato H, Sakai T, Sugaya T, Otaki Y, Aoki K, et al. (2009) Tocilizumab dramatically ameliorated life-threatening diarrhea due to secondary amyloidosis associated with rheumatoid arthritis. Clin Rheumatol 28: 1113-1116.
  115. Inoue D, Arima H, Kawanami C, Takiuchi Y, Nagano S, et al. (2010) Excellent therapeutic effect of tocilizumab on intestinal amyloid a deposition secondary to active rheumatoid arthritis. Clin Rheumatol 29: 1195-1197.
  116. Hattori Y, Ubara Y, Sumida K, Hiramatsu R, Hasegawa E, et al. (2012) Tocilizumab improves cardiac disease in a hemodialysis patient with AA amyloidosis secondary to rheumatoid arthritis. Amyloid 19: 37-40.
  117. Hakala M, Immonen K, Korpela M, Vasala M, Kauppi MJ (2013) Good medium-term efficacy of tocilizumab in DMARD and anti-TNF-α therapy resistant reactive amyloidosis. Ann Rheum Dis 72: 464-465.
  118. Okuda Y, Ohnishi M, Matoba K, Jouyama K, Yamada A, et al. (2013) Comparison of the clinical utility of tocilizumab and anti-TNF therapy in AA amyloidosis complicating rheumatic diseases. Mod Rheumatol .
  119. Kawashiri SY, Kawakami A, Yamasaki S, Imazato T, Iwamoto N, et al. (2011) Effects of the anti-interleukin-6 receptor antibody, tocilizumab, on serum lipid levels in patients with rheumatoid arthritis. Rheumatol Int 31: 451-456.
  120. Pradhan AD, Manson JE, Rifai N, Buring JE, Ridker PM (2001) C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA 286: 327-334.
  121. Fève B, Bastard JP (2009) The role of interleukins in insulin resistance and type 2 diabetes mellitus. Nat Rev Endocrinol 5: 305-311.
  122. Ogata A, Morishima A, Hirano T, Hishitani Y, Hagihara K, et al. (2011) Improvement of HbA1c during treatment with humanised anti-interleukin 6 receptor antibody, tocilizumab. Ann Rheum Dis 70: 1164-1165.
  123. Schultz O, Oberhauser F, Saech J, Rubbert-Roth A, Hahn M, et al. (2010) Effects of inhibition of interleukin-6 signalling on insulin sensitivity and lipoprotein (a) levels in human subjects with rheumatoid diseases. PLoS One 5: e14328.
  124. Hirao M, Yamasaki N, Oze H, Ebina K, Nampei A, et al. (2012) Serum level of oxidative stress marker is dramatically low in patients with rheumatoid arthritis treated with tocilizumab. Rheumatol Int 32: 4041-4045.
  125. Kume K, Amano K, Yamada S, Hatta K, Ohta H, et al. (2011) Tocilizumab monotherapy reduces arterial stiffness as effectively as etanercept or adalimumab monotherapy in rheumatoid arthritis: an open-label randomized controlled trial. J Rheumatol 38: 2169-2171.
  126. Protogerou AD, Zampeli E, Fragiadaki K, Stamatelopoulos K, Papamichael C, et al. (2011) A pilot study of endothelial dysfunction and aortic stiffness after interleukin-6 receptor inhibition in rheumatoid arthritis. Atherosclerosis 219: 734-736.
  127. Nishimoto N, Ito K, Takagi N (2010) Safety and efficacy profiles of tocilizumab monotherapy in Japanese patients with rheumatoid arthritis: meta-analysis of six initial trials and five long-term extensions. Mod Rheumatol 20: 222-232.
  128. Koike T, Harigai M, Inokuma S, Ishiguro N, Ryu J, et al. (2011) Postmarketing surveillance of tocilizumab for rheumatoid arthritis in Japan: interim analysis of 3881 patients. Ann Rheum Dis 70: 2148-2151.
  129. Gout T, Ostör AJ, Nisar MK (2011) Lower gastrointestinal perforation in rheumatoid arthritis patients treated with conventional DMARDs or tocilizumab: a systematic literature review. Clin Rheumatol 30: 1471-1474.
  130. Gómez-Reino JJ, Carmona L, Valverde VR, Mola EM, Montero MD; BIOBADASER Group. (2003) Treatment of rheumatoid arthritis with tumor necrosis factor inhibitors may predispose to significant increase in tuberculosis risk: a multicenter active-surveillance report. Arthritis Rheum 48: 2122-2127.
  131. Okada M, Kita Y, Kanamaru N, Hashimoto S, Uchiyama Y, et al. (2011) Anti-IL-6 receptor antibody causes less promotion of tuberculosis infection than anti-TNF-α antibody in mice. Clin Dev Immunol 2011: 404929.
  132. Ogata A, Mori M, Hashimoto S, Yano Y, Fujikawa T, et al. (2010) Minimal influence of tocilizumab on IFN-gamma synthesis by tuberculosis antigens. Mod Rheumatol 20: 130-133.
  133. Gallucci RM, Simeonova PP, Matheson JM, Kommineni C, Guriel JL, et al. (2000) Impaired cutaneous wound healing in interleukin-6-deficient and immunosuppressed mice. FASEB J 14: 2525-2531.
  134. Gallucci RM, Sugawara T, Yucesoy B, Berryann K, Simeonova PP, et al. (2001) Interleukin-6 treatment augments cutaneous wound healing in immunosuppressed mice. J Interferon Cytokine Res 21: 603-609.
  135. Lin ZQ, Kondo T, Ishida Y, Takayasu T, Mukaida N (2003) Essential involvement of IL-6 in the skin wound-healing process as evidenced by delayed wound healing in IL-6-deficient mice. J Leukoc Biol 73: 713-721.
  136. Lin TW, Cardenas L, Glaser DL, Soslowsky LJ (2006) Tendon healing in interleukin-4 and interleukin-6 knockout mice. J Biomech 39: 61-69.
  137. Yang X, Ricciardi BF, Hernandez-Soria A, Shi Y, Pleshko Camacho N, et al. (2007) Callus mineralization and maturation are delayed during fracture healing in interleukin-6 knockout mice. Bone 41: 928-936.
  138. Kuhn K, Miyoshi H, Manieri NA, Malvin NP, Bijanki V, et al. (2012) Anti-IL-6 therapy impairs intestinal repair through inhibition of epithelial proliferation after injury. Arthritis Rheum 64(Suppl 10): S710.
  139. McFarland-Mancini MM, Funk HM, Paluch AM, Zhou M, Giridhar PV, et al. (2010) Differences in wound healing in mice with deficiency of IL-6 versus IL-6 receptor. J Immunol 184: 7219-7228.
  140. Momohara S, Hashimoto J, Tsuboi H, Miyahara H, Nakagawa N, et al. (2012) Analysis of perioperative clinical features and complications after orthopaedic surgery in rheumatoid arthritis patients treated with tocilizumab in a real-world setting: results from the multicenter TOcilizumab in Perioperative Period (TOPP) study. Mod Rheumatol.
  141. Hirao M, Hashimoto J, Tsuboi H, Nampei A, Nakahara H, et al. (2009) Laboratory and febrile features after joint surgery in patients with rheumatoid arthritis treated with tocilizumab. Ann Rheum Dis 68: 654-657.
  142. Hiroshima R, Kawakami K, Iwamoto T, Tokita A, Yano K, et al. (2011) Analysis of C-reactive protein levels and febrile tendency after joint surgery in rheumatoid arthritis patients treated with a perioperative 4-week interruption of tocilizumab. Mod Rheumatol 21: 109-111.
  143. Terpos E, Fragiadaki K, Konsta M, Bratengeier C, Papatheodorou A, et al. (2011) Early effects of IL-6 receptor inhibition on bone homeostasis: a pilot study in women with rheumatoid arthritis. Clin Exp Rheumatol 29: 921-925.
  144. Kanbe K, Nakamura A, Inoue Y, Hobo K (2012) Osteoprotegerin expression in bone marrow by treatment with tocilizumab in rheumatoid arthritis. Rheumatol Int 32: 2669-2674.
  145. Tanaka T, Narazaki M, Kishimoto T (2012) Therapeutic targeting of the interleukin-6 receptor. Annu Rev Pharmacol Toxicol 52: 199-219.
  146. Tanaka T, Kishimoto T (2012) Targeting interleukin-6: all the way to treat autoimmune and inflammatory diseases. Int J Biol Sci 8: 1227-1236.
  147. Samson M, Audia S, Janikashvili N, Ciudad M, Trad M, et al. (2012) Brief report: inhibition of interleukin-6 function corrects Th17/Treg cell imbalance in patients with rheumatoid arthritis. Arthritis Rheum 64: 2499-2503.
  148. Pesce B, Soto L, Sabugo F, Wurmann P, Cuchacovich M, et al. (2013) Effect of interleukin-6 receptor blockade on the balance between regulatory T cells and T helper type 17 cells in rheumatoid arthritis patients. Clin Exp Immunol 171: 237-242.
  149. Carbone G, Wilson A, Diehl SA, Bunn J, Cooper SM, et al. (2013) Interleukin-6 receptor blockade selectively reduces IL-21 production by CD4 T cells and IgG4 autoantibodies in rheumatoid arthritis. Int J Biol Sci 9: 279-288.
  150. Roll P, Muhammad K, Schumann M, Kleinert S, Einsele H, et al. (2011) In vivo effects of the anti-interleukin-6 receptor inhibitor tocilizumab on the B cell compartment. Arthritis Rheum 63: 1255-1264.
  151. Muhammad K, Roll P, Seibold T, Kleinert S, Einsele H, et al. (2011) Impact of IL-6 receptor inhibition on human memory B cells in vivo: impaired somatic hypermutation in preswitch memory B cells and modulation of mutational targeting in memory B cells. Ann Rheum Dis 70: 1507-1510.
  152. Smolen JS, Landewé R, Breedveld FC, Dougados M, Emery P, et al. (2010) EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs. Ann Rheum Dis 69: 964-975.
  153. Singh JA, Furst DE, Bharat A, Curtis JR, Kavanaugh AF, et al. (2012) 2012 update of the 2008 American College of Rheumatology recommendations for the use of disease-modifying antirheumatic drugs and biologic agents in the treatment of rheumatoid arthritis. Arthritis Care Res (Hoboken) 64: 625-639.
  154. Smolen JS, Schoels MM, Nishimoto N, Breedveld FC, Burmester GR, et al. (2013) Consensus statement on blocking the effects of interleukin-6 and in particular by interleukin-6 receptor inhibition in rheumatoid arthritis and other inflammatory conditions. Ann Rheum Dis 72: 482-492.
  155. Schoels MM, van der Heijde D, Breedveld FC, Burmester GR, Dougados M, et al. (2013) Blocking the effects of interleukin-6 in rheumatoid arthritis and other inflammatory rheumatic diseases: systematic literature review and meta-analysis informing a consensus statement. Ann Rheum Dis 72: 583-589.
  156. Bergman GJ, Hochberg MC, Boers M, Wintfeld N, Kielhorn A, et al. (2010) Indirect comparison of tocilizumab and other biologic agents in patients with rheumatoid arthritis and inadequate response to disease-modifying antirheumatic drugs. Semin Arthritis Rheum 39: 425-441.
  157. Salliot C, Finckh A, Katchamart W, Lu Y, Sun Y, et al. (2011) Indirect comparisons of the efficacy of biological antirheumatic agents in rheumatoid arthritis in patients with an inadequate response to conventional disease-modifying antirheumatic drugs or to an anti-tumour necrosis factor agent: a meta-analysis. Ann Rheum Dis 70: 266-271.
  158. Schoels M, Aletaha D, Smolen JS, Wong JB (2012) Comparative effectiveness and safety of biological treatment options after tumour necrosis factor? Inhibitor failure in rheumatoid arthritis: systematic review and indirect pairwise meta-analysis. Ann Rheum Dis 71: 1303-1308.
  159. Singh JA, Wells GA, Christensen R, Tanjong Ghogomu E, Maxwell L, et al. (2011) Adverse effects of biologics: a network meta-analysis and Cochrane overview. Cochrane Database Syst Rev : CD008794.
  160. Klareskog L, van der Heijde D, de Jager JP, Gough A, Kalden J, et al; TEMPO (Trial of Etanercept and Methotrexate with Radiographic Patient Outcome) study investigators (2004) Therapeutic effect of the combination of etanercept and methotrexate compared with each treatment alone in patients with rheumatoid arthritis: double-blind randomised controlled trial. Lancet 363: 675-681.
  161. Breedveld FC, Weisman MH, Kavanaugh AF, Cohen SB, Pavelka K, et al. (2006) The PREMIER study: A multicenter, randomized, double-blind clinical trial of combination therapy with adalimumab plus methotrexate versus methotrexate alone or adalimumab alone in patients with early, aggressive rheumatoid arthritis who had not had previous methotrexate treatment. Arthritis Rheum 54: 26-37.
  162. Keystone EC, Genovese MC, Klareskog L, Hsia EC, Hall ST, et al. (2009) Golimumab, a human antibody to tumour necrosis factor ? given by monthly subcutaneous injections, in active rheumatoid arthritis despite methotrexate therapy: the GO-FORWARD Study. Ann Rheum Dis 68: 789-796.
  163. Gabay C, Emery P, van Vollenhoven R, Dikranian A, Alten R, et al. (2013) Tocilizumab monotherapy versus adalimumab monotherapy for treatment of rheumatoid arthritis (ADACTA): a randomised, double-blind, controlled phase 4 trial. Lancet 381: 1541-1550.
  164. Ohta S, Tsuru T, Terao K, Mogi S, Suzaki M, et al. (2010) A phase I/II study evaluating the safety, pharmacokinetics and clinical response of tocilizumab for subcutaneous administration in patients with rheumatoid arthritis. Ann Rheum Dis 69(Suppl 3): 543.
  165. Ogata A, and MUSASHI Study Group (2012) The MUSASHI study: comparison of subcutaneous tocilizumab monotherapy versus intravenous tocilizumab monotherapy: results from a double-blind, parallel-group, comparative phase III non-inferiority study in Japanese patients with rheumatoid arthritis. Ann Rheum Dis 71(Suppl 3): 373.
  166. Burmester GR, Rubbert-Roth A, Cantagrel AG, Hall S, Leszczynski P, et al. (2012) A randomized, double-blind, parallel group study of the safety and efficacy of tocilizumab SC versus tocilizumab IV, in combination with traditional DMARDs in patients with moderate to severe RA. Arthritis Rheum 64 (Suppl 10): S1075.
  167. Kivitz AJ (2012) A randomized, double-blind, parallel-group study of the safety and efficacy of tocilizumab subcutaneous versus placebo in combination with traditional DMARDs in patients with moderate to severe rheumatoid arthritis (BREVACTA). Presented at ACR2012 Washington DC Late-Breaking Abstracts L8.
  168. Igawa T, Ishii S, Tachibana T, Maeda A, Higuchi Y, et al. (2010) Antibody recycling by engineered pH-dependent antigen binding improves the duration of antigen neutralization. Nat Biotechnol 28: 1203-1207.
  169. Jones SA, Scheller J, Rose-John S (2011) Therapeutic strategies for the clinical blockade of IL-6/gp130 signaling. J Clin Invest 121: 3375-3383.
  170. Huizinga TW, Kivitz AJ, Rell-Bakalarska M, Fleischmann RM, Jasson M, et al. (2012) Sarilumab for the treatment of moderate to severe rheumatoid arthritis: Results of a phase 2, randomized double blind placebo controlled international study. Ann Rheum Dis 71(Suppl 3): 60.
  171. Roy MV, Ulrichts H, Rossenu S, Jacobs S, Poelmans S, et al. (2012) Preclinical development of ALX-0061, an anti-IL-6R nanobody(R) for therapeutic use in rheumatoid arthritis with a high in vitro affinity and potency and a competitive in vivo pharmacological profile. Arthritis Rheum 64 (Suppl 10): S146.
  172. Xu Z, Bouman-Thio E, Comisar C, Frederick B, Van Hartingsveldt B, et al. (2011) Pharmacokinetics, pharmacodynamics and safety of a human anti-IL-6 monoclonal antibody (sirukumab) in healthy subjects in a first-in-human study. Br J Clin Pharmacol 72: 270-281.
  173. Zhuang Y, Xu Z, de Vries DE, Wang Q, Shishido A, et al. (2013) Pharmacokinetics and safety of sirukumab following a single subcutaneous administration to healthy Japanese and Caucasian subjects. Int J Clin Pharmacol Ther 51: 187-199.
  174. Hsu B, Sheng S, Weinblatt ME, Smolen JS (2012) Results from a multicenter, international, randomized, double blind, placebo controlled, phase 2 study of Sirukumab, a human anti-IL-6 monocloncal antibody, in patients with active rheumatoid arthritis despite methotrexate therapy. Ann Rheum Dis 71(Suppl 3): 61.
  175. Hsu B, Sheng S, Smolen JS, Weinblatt ME (2012) Results from a 2-part, proof of concept, dose ranging, randomized, double blind, placebo controlled, phase 2 study of Sirukumab, a human anti-IL-6 monoclonal antibody, in patients with active rheumatoid arthritis despite methotrexate therapy. Ann Rheum Dis 71(Suppl 3): 188.
  176. Mease P, Strand V, Shalamberidze L, Dimic A, Raskina T, et al. (2012) A phase II, double-blind, randomised, placebo-controlled study of BMS945429 (ALD518) in patients with rheumatoid arthritis with an inadequate response to methotrexate. Ann Rheum Dis 71: 1183-1189.
  177. Hickling M, Golor G, Jullion A, Shaw S, Kretsos K (2011) Safety and pharmacokinetics of CDP6038, an anti-IL-6 monoclonal antibody, administered by subcutaneous injection and intravenous infusion to health male volunteers: a phase 1 study. Ann Rheum Dis 70 (Suppl 3): 471.
  178. Finch DK, Sleeman MA, Moisan J, Ferraro F, Botterell S, et al. (2011) Whole-molecule antibody engineering: generation of a high-affinity anti-IL-6 antibody with extended pharmacokinetics. J Mol Biol 411: 791-807.
  179. Tenhumberg S, Waetzig GH, Chalaris A, Rabe B, Seegert D, et al. (2008) Structure-guided optimization of the interleukin-6 trans-signaling antagonist sgp130. J Biol Chem 283: 27200-27207.
  180. Yokota S, Tanaka T, Kishimoto T (2012) Efficacy, safety and tolerability of tocilizumab in patients with systemic juvenile idiopathic arthritis. Ther Adv Musculoskelet Dis 4: 387-397.
  181. Tanaka T, Ogata A, Shima Y, Narazaki M, Kumanogoh A, et al. (2012) Therapeutic implications of tocilizumab, a humanized anti-interleukin-6 receptor antibody, for various immune-mediated diseases: an update review. Curr Rheumatol Rev 8: 209-226.
  182. Sieper J, Porter-Brown B, Thompson L, Harari O, Dougados M (2012) Tocilizumab (TCZ) is not effective for the treatment of ankylosing spondylitis (AS): Results of a phase 2, international, multicenter, randomised, double-blind, placebo-controlled trial. Ann Rheum Dis 71: 110.
  183. Sieper J, Inman RD, Badalamenti S, Radin A, Braun J (2012) Sarilumab for the treatment of ankylosing spondylitis: Results of a phase 2, randomized, double-blind, placebo-controlled, international study (ALIGN). Ann Rheum Dis 71: 111.
Citation: Tanaka T, Ogata A, Kishimoto T (2013) Targeting of Interleukin-6 for the Treatment of Rheumatoid Arthritis: A Review and Update. Rheumatol Curr Res S4:002.

Copyright: © 2013 Tanaka T, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top