81
Views
7
CrossRef citations to date
0
Altmetric
Review

Non-tumor necrosis factor-based biologic therapies for rheumatoid arthritis: present, future, and insights into pathogenesis

&
Pages 1-12 | Published online: 09 Dec 2013

Abstract

The way rheumatoid arthritis is treated has changed dramatically with the introduction of anti-tumor necrosis factor (anti-TNF) biologics. Nevertheless, many patients still have less than adequate control of their disease activity even with these therapeutic regimens, and current knowledge fails to explain all the data already gathered. There is now a wide range of drugs from different classes of biologic disease-modifying anti-rheumatic drugs available (and soon this number will increase significantly), that provides the opportunity to address each patient as a particular case and thereby optimize medical intervention. Currently available biologics for the treatment of rheumatoid arthritis apart from anti-TNF-based therapies are reviewed, along with an analysis of the new insights they provide into the pathogenesis of the disease and a discussion of future prospects in the area.

Introduction

Rheumatoid arthritis (RA) is a chronic, systemic, inflammatory disease of adulthood. Although both sexes and every age group may be affected, it is more frequent in women in the fourth and fifth decades of life. The disease may cause damage to various organs and tissues, including the lungs, skin, and heart, but predominantly affects the joints. Further, there is a strong association with increased cardiovascular risk, which ultimately leads to premature death.Citation1 Arthritis is the most recognized and studied manifestation of the disease, being the area in which the most extensive data have been gathered from a pathophysiologic point of view, and the one that is evaluated in the large majority of clinical trials.

The way RA is treated has changed dramatically in the last 15 years with the introduction of biologics, namely anti-tumor necrosis factor (TNF) therapy. With the use of these molecular targeted therapies, the standard of care for patients with RA has improved, better clinical results have been achieved, and knowledge about the pathogenesis of RA has grown exponentially. Nevertheless, many patients still have less than adequate control of their disease activity even with these therapeutic regimens or become resistant to their beneficial effects. This imposes the need for other treatment options, and suggests that current theories about RA may be only partially correct. In recent years, other drugs have been produced that target other components of the inflammatory process in RA, with good efficacy and safety profiles, which eventually led to their approval and widespread use. It is the aim of this paper to review the biologics currently available for the treatment of RA apart from anti-TNF-based therapies, to analyze the new insights they provide into the pathogenesis of the disease, and to discuss future prospects in the area.

Etiology and pathogenesis

A clear etiology for RA has never been identified. Genetic factors clearly play a role, as revealed by an up to 30% concordance rate in monozygotic twins,Citation2,Citation3 and most genomic association studies point to the major histocompatibility complex locus as the one with the highest relative risk.Citation4 The most important environmental risk factor is smoking, and some microbes (such as Porphyromonas gingivalis infection in periodontal tissue) have been implicated, but this has been regarded as of uncertain value.Citation5

RA is characterized by activation of resident synovial inflammatory cells, mainly macrophages, infiltration of lymphocytes and neutrophils in the synovium, and the production of an inflammatory milieu, which in turn promotes proliferation of synoviocytes and fibroblasts, and neoangiogenesis. The presence of autoantibodies (rheumatoid factor and anti-cyclic citrullinated peptide antibodies), a reflection of the role of B-cells, is one of the laboratory hallmarks of RA, in some cases being detected more than 10 years before clinical onset.Citation6 All this leads to an aberrant, hyperplastic architecture of the synovial membrane, the rheumatoid pannus, and to the differentiation and activation of osteoclasts and subsequent bone destruction. Chondrocyte function is also altered, leading to degradation of cartilage and biomechanical derangement of normal articular function. Periarticular structures such as ligaments and tendons are also eventually involved in the inflammatory process, culminating in further dysfunction and production of the typical clinical and radiologic picture of RA.

As such, apart from macrophages and other “effector” cell types (dendritic cells, neutrophils, synoviocytes, osteo-blasts, osteoclasts, and chondrocytes), three components of RA pathogenesis have become recognized as major players based on both basic and clinical research, ie, B-cells, T-cells, and a wide range of inflammatory cytokines and growth factors that, acting as an intricate and redundant network both systemically and locally, shift the balance towards a proinflammatory state. Accumulating evidence shows that all these players act interdependently, and have continuously challenged our understanding of immune physiology and pathology.

After TNF blockers were introduced in the clinical management of RA, two types of clinical picture have emerged in daily practice, ie, an inadequate response and/or existence of contraindications or intolerance, precluding the use of these agents and raising the need to find alternatives. In fact, anti-TNF therapy achieves a 20% improvement in American College of Rheumatology response criteria (ACR20) in about 42%–85% of patients, and an ACR50 response in only 21%–69%,Citation7,Citation8 with secondary failure rates of up to 50% during the first year.Citation9

Current molecular targeted strategies to control RA (beyond TNF) have tried to block at least one of the three components mentioned, and the most relevant of those are reviewed here.

B-cell-targeted therapies

Role of B-cells in RA

Improvement in RA through B-cell depletion has highlighted the importance of B-cells in the pathogenesis of the disease. The presence of rheumatoid factor is related to disease severity and the frequency of extra-articular manifestations,Citation10 and anti-cyclic citrullinated peptide antibodies are related to aggressiveness of the disease.Citation11 Moreover, baseline rheumatoid factor seropositivity seems to be related to the response to rituximab.Citation12Citation14 However, CD20 is lacking in antibody-producing plasmablasts and plasma cells; the response to rituximab is related to the level of B-cell depletion in peripheral bloodCitation15,Citation16 and synovial tissue,Citation17 and is coincident with a reduction in the number of peripheral memory B-cells (CD19+/CD27+),Citation18 and not with the degree of reduction in plasma immunoglobulins.Citation19 Further, relapse is also related to B-cell repopulation,Citation20,Citation21 and non-antibody-producing B-cells are able to activate T-cells and produce articular disease.Citation22 All this reinforces the idea of an important role of B-cells beyond antibody production.

B-cells are potent antigen-presenting cellsCitation22 in the context of multiple diseases.Citation23,Citation24 They are able to activate CD4+ T-cells, and their presence is necessary for T-cell activation in synovial tissue.Citation25 B-cells are also capable of enhancing the differentiation of T-cells into the inflammatory T-helper (Th)17 phenotype.Citation26 Further, B-cells are potent cytokine producers that act not only in an autocrine manner (interleukin [IL]-10) but also activate other immune cells, including macrophages, neutrophils, and dendritic cells (IL-6, TNF-α, IL-1, and IL-10).Citation27 They are also important sources of potent chemotactic molecules that are crucial to pannus development.Citation28,Citation29 These findings add a twist to the classical view of T-helper cells ensuring activation and maturation of B-cells and innate immunity activating adaptive immunity. They also suggest that the presence of autoantibodies may be interpreted in RA as a manifestation of loss of tolerance and of the presence of autoreactive B-cells that are themselves pathogenic, even before differentiating into permanent antibody-producing plasma cells.

The case for rituximab

Blocking the contribution of B-cells to disease activity has been achieved by B-cell-depleting therapies with great success. Rituximab, a chimeric monoclonal anti-CD20 antibody originally used in non-Hodgkin’s lymphoma, induces antibody-dependent cell-mediated cytotoxicity, complement-dependent cytotoxicity, and apoptosis of B-cells in various stages of development,Citation27,Citation30,Citation31 leading to their transient but almost complete depletion in peripheral blood, although only partially in the bone marrow and synovial tissue niches.Citation32,Citation33

The beneficial effect of rituximab-induced B-cell depletion has been substantiated in various major randomized clinical trials in RA, as well as in other immune-mediated diseases.Citation34 The benefit is reflected in a reduction of clinical disease activity and laboratory inflammation markers, halted radiographic progression, and better functional ability and quality of life scores. The effect consistently lasts for more than 6 months after a single course of two administrations of the drug, separated by 15 days, with clear benefit when compared with placebo, and with a good safety profile. This has been observed in methotrexate-naïve patients (anti-TNF-naïve) in IMAGE (International Study in Methotrexate-naïve Subjects Investigating Rituximab’s Efficacy),Citation13 and in methotrexate-inadequate responders in SERENECitation40 (Study Evaluating Rituximab’s Efficacy in MTX iNadequate rEsponders) and MIRRORCitation35 (Methotrexate Inadequate Responders Randomized study Of Rituximab), with comparable results. In SERENE,Citation40 ACR20, ACR50, and ACR70 responses at 24 weeks were 51%, 26%, and 10%, respectively, with a clear-cut difference from placebo (23%, 9%, and 5%). Importantly, in REFLEXCitation36 (Randomized Evaluation oF Long-term Efficacy of rituXimab in RA), rituximab again produced similar results in anti-TNF-inadequate responders, with ACR20, ACR50, and ACR70 responses at 24 weeks of 51%, 27%, and 12%, respectively. A benefit from placebo has been shown for up to 5 years of continued rituximab retreatment in a post hoc open-label extension of REFLEX.Citation37 The MIRAR trial focused on an important issue directly comparing the use of an alternative anti-TNF versus rituximab after a first anti-TNF failure, with both treatments producing comparable benefits.Citation38 Conversely, several observational studies have suggested that rituximab may be more effective than a second anti-TNF in some subgroups,Citation39 although clinical trials confirming this are lacking.

Rituximab has shown to be effective in monotherapy, but considerably less so when used in combination with methotrexate, both in the magnitude and duration of benefit.Citation12,Citation40 This is one of the major caveats of using rituximab in RA because tocilizumab is still preferred as an alternative to anti-TNF agents for methotrexate-intolerant patients or when methotrexate is contraindicated, such as in severe pulmonary disease. The use of rituximab with other synthetic disease-modifying antirheumatic drugs (DMARDs), mainly leflunomide, has been evaluated as producing similar results,Citation41 but large head-to-head randomized trials are still lacking.

In light of these accumulated data, there seems to be no reason for rituximab not being recommended as a first-line agent. However, rituximab’s debut in the treatment of RA was somewhat obscured by the initial enthusiasm for anti-TNF agents, and its initial use in malignancies may have raised some concern regarding its widespread use. In Europe, rituximab was approved in RA patients with severe disease, but only after failure of at least one anti-TNF agent (and always in association with methotrexate).Citation42 However, in the more recent 2012 ACR guidelines, rituximab has already been recommended as a first-line biologic agent after synthetic DMARD failure for patients with moderate to severe disease activity and for those with low disease activity and poor prognosis markers.Citation43

Some controversy still exists regarding the optimal dosage of rituximab. The evidence suggests that a 2 × 500 mg regimen has almost similar efficacy to the 2 × 1 g regimen used in the majority of trials, with a reduced rate of infusion reactions and adverse events.Citation44 However, the full-dose regimen seems to result in better ACR70 response rates, and better radiographic outcomes were produced in the IMAGE trial.Citation13 As such, the approved dose in most consensus guidelines is 2 × 1 g.

Beyond rituximab

CD20 is absent in long-lived plasma cells and plasmablasts and is not expressed until after the pre-B stage of B-cell development. As such, rituximab therapy is not expected to interfere directly with permanent antibody-producing cells, but rather with their precursors. Also, the bone marrow consists of a protective niche, being largely refractory to B-cell depletion after administration of rituximab.Citation32,Citation33,Citation45 Importantly, synovial tissue seems to be also relatively resistant to the action of rituximab.Citation32,Citation33 In RA, the synovial tissue is infiltrated with B-cells and T-cells, producing three types of synovitis: diffuse synovitis, in which both types of cells are interspersed with no apparent spatial organization; follicular synovitis, in which ectopic germinal centers form, with a nucleus of B-cells surrounded by T-cells and with autoantibody-secreting capacity;Citation46 and granulomatous synovitis. Sex, age, disease duration, and previous treatment do not correlate with the type of synovitis found, and each patient shows only one type. This suggests that there may be different subtypes of the disease. Accordingly, seronegative RA patients tend to have a diffuse pattern, and all patients with follicular synovitis were seropositive in a cohort of 21 cases.Citation47 Additionally, rheumatoid nodules, a common extra-articular manifestation of the disease, seem to be closely associated with granulomatous synovitis. Interestingly, rheumatoid factor levels tend to be higher and the effect of rituximab on immunoglobulin levels in synovial fluid tends to be more pronounced in follicular synovitis.Citation32 In one study, the diameter of follicular aggregates was significantly associated with disease severity,Citation48 but no comparison was made between the different types of synovitis. The clinical efficacy of rituximab or even baseline disease activity in each subgroup was never evaluated. Nevertheless, these studies show that B-cells in local synovial tissue are crucial determinants of the disease, and a more potent way of inhibiting B-cell function in the synovium could be interesting.

CD19, a membrane-associated molecule involved in modulation of the threshold of B-cell activation, is present in a larger range of B-cells, including plasmablasts, plasma cells, and pre-B cells,Citation49 and is also specific for B-cells. In contrast with rituximab, administration of anti-CD19 monoclonal antibody depletes the majority of B-cells in the bone marrow, lymphoid organs, and synovial tissue,Citation50 as well as some anti-CD20-resistant subtypes resident in the peritoneal cavity (B1a, B1b),Citation50,Citation51 potentially leading to an increased clinical efficacy, as was observed in animal models of RA. There is, however, some concern that depletion of regulating B10 (CD20-) cells could induce autoimmunity.Citation52 Depletion of plasma cells also induces at least a partial loss of protective immunity, which could lead to increased safety issues concerning the risk of infection. These issues have not been fully addressed in humans, with the only experience having been gathered in patients with hematologic malignancies, first with a mouse anti-hCD19,Citation53 then with a humanized version,Citation54 with both having shown an acceptable safety profile.

Other strategies to block the B-cell contribution to RA have been envisaged, including blocking B-cell activation with an anti-BAFF/BLyS (B-cell activating factor/B-lymphocyte stimulator) antibody (belimumab)Citation55 or with a TACI-immunoglobulin fusion protein (atacicept) which blocks both BLyS and APRIL (A PRoliferation Inducing Ligand) costimulators.Citation56 Belimumab has shown modest efficacy (only ACR20 responses different from placebo), although with a good safety profile, mainly in moderate to severe, seropositive, anti-TNF-naïve RA patients who have failed methotrexate therapy.Citation57 Atacicept did not meet expectations, producing no clinical improvement, but is safe to use in methotrexate-inadequate respondersCitation58 or after antiTNF failure.Citation56 It is interesting to note that atacicept fulfilled its expected biological effect,Citation59 reducing circulating immunoglobulin levels and autoantibody titers,Citation58 which reinforces the notion of autoantibodies being markers of disease and not effectors per se.

T-cell-targeted therapies

Role of T-cells in RA

T-cells are found within the synovial tissue in RA, and play a major role in perpetuating inflammation and producing tissue damage. They are activated by antigen-presenting cells, including dendritic cells, macrophages, and even B-cells, and subsequently secrete various cytokines (which contribute to inflammation in synovial tissue), activate innate immune cells, support B-cell activation, and induce destructive chondrocyte and osteoclast activation. Various subtypes of T-helper cells have been described, and modulation of the differentiation of naïve T-cells into each of those types is being increasingly studied, and proving to be central in RA.

The inflammatory pattern in RA has long been characterized by a shift towards a Th1 phenotype in CD4+ T-cell differentiation. This leads to a predominant production of Th1-associated cytokines such as interferon-gamma, with Th2-associated cytokines such as IL-4 and IL-13 found to be nearly absent in the synovial tissue.Citation60 The Th17 subtype of CD4+ T-cells is today recognized as a major player in synovial inflammation and bone erosion, mainly by the production of IL-17 and IL-23.Citation61 In fact, the pivotal role of Th1 in the pathogenesis of RA has been increasingly recognized as taking place via actions ascribed to Th17, and it is not clear whether Th17 cells consist of a specialized Th1 subtype or a different phenotype that would act in concert with Th1 cells. Th17 differentiation from naïve T-cells seems to require IL-6 and transforming growth factor-beta concomitant signaling, and their proliferation is dependent on IL-23, produced by antigen-presenting cells.Citation62,Citation63 In any case, both subtypes seem to have an important role in the regulation of inflammation in RA. The relative weight of each one is still under discussion.

Conversely, inhibition of regulatory T-cell (Treg) differentiation seems to be important in the perpetuation of autoimmune diseases,Citation64 and their numbers and activation status are associated with RA activity in generalCitation65,Citation66 and with the response to treatment with either synthetic or biologic DMARDs.Citation67,Citation68

Case for abatacept

Efforts to block T-cell activity in RA have produced important results.Citation69 Abatacept, a human CTLA4-IgFc (CTLA, cytotoxic T-lymphocyte antigen) fusion protein, binds to CD80/86 on antigen-presenting cells and competitively prevents its binding to the T-cell CD28 molecule (CTLA). CD80/CD86-CD28 interaction produces a crucial costimulatory signal upon major histocompatibility complex-T-cell receptor interaction, and its blocking inhibits T-cell activation. Administration of abatacept subsequently leads to impaired B-cell activation and reduced levels of autoantibodies, together with a reduction in T-cell-mediated activation of osteoclasts and diminished cytokine release from T-cells, B-cells, and macrophages.Citation70,Citation71 This translates into reduced disease activity, a decrease in inflammatory markers and halted bone erosion, as seen in various randomized clinical trials supporting its use, with benefits being present as early as 1–4 months of continued administration.Citation72 Abatacept is more effective in anticyclic citrullinated peptide antibody-positive patients.Citation73 The reason for this is unknown, and emphasizes that RA is a syndrome with multiple subtypes that may respond differently to different therapies.

The efficacy and safety of abatacept was consistently shown in biologic-naïve methotrexate-inadequate responders in the AIM (Abatacept in Inadequate responders to Methotrexate) trial,Citation74 with ACR20, ACR50, and ACR70 responses of 68%, 40%, and 20%, respectively, compared with 40%, 17%, and 6.5% with placebo, at 24 weeks. In the same group of patients, abatacept produced similar results as a first-line biologic agent when compared with the anti-TNF agent infliximab in ATTEST (Abatacept or infliximab versus placebo, a Trial for Tolerability, Efficacy and Safety in Treating RA).Citation75 In this trial, it was even suggested that abatacept could be more effective than infliximab in sustaining clinical improvement and remission rates after 12 months of therapy.

Abatacept also proved to be an effective therapeutic option after failure of an anti-TNF agent in ATTAIN (Abatacept Trial in Treatment of Anti-TNF INadequate responders)Citation76 (ACR20, ACR50, and ACR70 responses of 50%, 20%, and 10%, respectively, versus 19%, 4%, and 1% in controls), and also in the open-label extension of the ATTEST trial, with infliximab-inadequate responders being rescued by abatacept.Citation77 Additionally, no washout period is necessary before abatacept treatment after anti-TNF failure.Citation78

Because clinical guidelines increasingly value the recognition of poor prognostic markers to justify the immediate beginning of biologic DMARDs, abatacept also produced a good clinical and radiographic response in methotrexate-naïve patients with early erosive RA in AGREE (Abatacept trial to Gauge Remission and joint damage progression in methotrexate-naïve patients with Early Erosive rheumatoid arthritis).Citation79

Administration of abatacept in the majority of clinical trials refers to the 20 mg/kg monthly intravenous regimen, on background methotrexate. It has become increasingly clear that subcutaneous administration is equally effective and safe.Citation80 Subcutaneous injection requires a much smaller dose (125 mg weekly injections, 500 mg per month), which has a clear advantage when considering cost-effectiveness.

Abatacept monotherapy (without methotrexate) has not been sufficiently studied. In the ACCOMPANY (Abatacept in Subjects with Rheumatoid Arthritis Administered Plus or Minus Background Methotrexate Subcutaneously) trial, subcutaneous abatacept showed similar efficacy and immunogenicity whether with or without methotrexate.Citation81 Data regarding combination regimens with other synthetic DMARDs are sparse.

Abatacept is presently included in clinical consensus and guidelines in Europe as an option after anti-TNF failure, always in combination with methotrexate.Citation42 In Switzerland, it has already been adopted as a first-line biologic agent, in concordance with the results referred to above.Citation72 In the US, like rituximab, it has also been included in the recommendation guidelinesCitation43 as a first-line biologic agent for all patients with moderate to severe disease activity and for patients with low activity and poor prognostic markers who have failed synthetic DMARDs.

Beyond abatacept

Abatacept, as a costimulation blocker, inhibits the activation and proliferation of all T-cell subtypes,Citation82 including Tregs. This is in contrast with the mechanism of action of other biologic agents such as anti-IL-6R, anti-CD20, or anti-TNF, which enhance Treg regulatory function, thereby ameliorating disease. Maintaining Treg function by selectively inhibiting other T-cell subtypes would imply a different molecular target, as CTLA-IgFc is nonspecific. More precise modulation of the T-cell activation pattern, selectively inhibiting Th1 and Th17 subtypes, would theoretically result in a more effective way of controlling disease activity, maybe even with fewer adverse effects with respect to susceptibility to infection.

Combination therapy with other biologics has been tried. Association with an anti-TNF agent (etanercept) did not produce significant clinical benefits, but induced a serious rise in adverse event rates.Citation83 Combination regimens with other biologics such as tocilizumab remain to be tested.

Anti-cytokine agents

Targeting specific cytokines involved in the inflammation process in the joints and systemically has been a logical strategy for controlling disease activity.Citation84 It modulates not only the direct effects of these cytokines on critical “effector” cells such as osteoclasts, chondrocytes, or macrophages, but also has indirect cell-mediated effects subsequent to the differential pattern of activation and differentiation of T-cells and B-cells.Citation85 Anti-TNF agents were the first successful case, leading to the most impressive improvement in the care of patients with RA. Targeting IL-6 signaling was the next successful step, leading to widespread use of the anti-IL-6 agent tocilizumab. In the future, drugs inhibiting the signaling of other cytokines such as IL-12, IL-13, and IL-17 may be promising strategies in the treatment of RA.

IL-6 in RA

IL-6 is produced by several cell types, including dendritic cells, B-cells, macrophages, and endothelial cells, upon their induction by inflammatory stimuli such as TNF-α, and is present in large quantities in the synovial fluid of RA-affected joints and in serum. It has a central role in the pathogenesis of RA, inducing endothelial activation, neutrophil activation and chemotaxis, directly activating osteoclasts and chondrocytes and stimulating matrix turnover and bone erosion.Citation86 IL-6 is also responsible for important biases in T-cell development.Citation62 Through inhibition of interferon-gamma and enhancing IL-4 signaling, IL-6 produces a shift towards a Th2 phenotype of CD4+ T-cells, which is in contrast with the main Th1 response seen in RA. An explanation for this contradiction may come from the discovery that IL-6 is central to the Th17 differentiation of naïve T-cells when in the presence of transforming growth factor-beta.Citation62 Th17 T-cells are known to be potent inflammatory cells in the synovium, mainly through the production of IL-17, which inhibits Th2 differentiation in favor of the Th1 phenotype; this could explain the contradictory isolated actions of IL-6, which also inhibits Treg differentiation, contributing to inflammation and loss of tolerance.

Further, IL-6 is largely responsible for the systemic effects of RA.Citation87 By inducing the expression of hepcidin, IL-6 contributes to the inflammatory anemia seen in many patients. Fatigue in RA is closely correlated with IL-6 concentrations in serum. IL-6 induces the production of acute-phase reactants in hepatocytes via the JAK/STAT signaling pathway, including C-reactive protein and serum amyloid A. C-reactive protein binds to apolipoprotein B-containing lipoproteinsCitation88 and increases the uptake of low-density lipoprotein in atheromatous lesions.Citation89 Serum amyloid A, in turn, was shown to be introduced into cholesterol-containing high-density lipoprotein particles, impairing protective reverse cholesterol transportCitation90,Citation91 and inducing formation of foam cells.Citation92

The case for tocilizumab

Blocking IL-6 signaling by administration of tocilizumab has produced consistent results whether in combination with methotrexate or in monotherapy.Citation93 In OPTIONCitation94 (Tocilizumab Pivotal Trial in Methotrexate Inadequate RespONders), the superiority of tocilizumab with methotrexate was shown in methotrexate-inadequate responders, when compared with methotrexate alone, for both clinical response and inflammatory markers (ACR20, ACR50, and ACR70 responses of 59%, 44%, and 22%, respectively, at 24 weeks versus 26%, 22%, and 4% with placebo). The LITHE (Tocilizumab Safety and the Prevention of Structural Joint Damage)Citation95 study confirmed a benefit in radiographic outcomes for tocilizumab. In the TOWARDCitation96 (Tocilizumab in Combination With Traditional DMARD Therapy) trial, a significant response was also seen when in combination with any synthetic DMARD versus any DMARD in monotherapy in patients with an inadequate response to synthetic DMARD therapy. After failure of one anti-TNF agent, tocilizumab also proved to be beneficial in combination with methotrexate (ACR20, ACR50, and ACR70 responses of 50%, 29%, and 12% at 24 weeks versus 10%, 3.8%, and 1.3% with methotrexate only) in the RADIATECitation97 (Research on Tocilizumab Determining Efficacy after Anti-TNF Failures) trial.

The efficacy of tocilizumab in monotherapy has been increasingly substantiated. In the ACT-RAYCitation98 (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) study, addition of methotrexate to tocilizumab therapy in methotrexate-inadequate responders brought no benefit, and the AMBITIONCitation99 (Comparison of tocilizumab monotherapy versus methotrexate monotherapy in patients with moderate to severe rheumatoid arthritis) trial showed clear superiority of tocilizumab in monotherapy versus methotrexate alone as soon as 6 months in patients naïve to any biologic or methotrexate. These data make tocilizumab an excellent choice (if not the only one) in the large group of patients who are intolerant or have contraindications to methotrexate (about 30%).Citation100

An important advantage of tocilizumab is its ability to control the extra-articular manifestations of RA, namely chronic inflammatory anemia, asthenia, and cardiovascular risk. Tocilizumab was seen to raise hemoglobin levels and to alleviate asthenia.Citation87 Initially, the impact of tocilizumab on the lipid profile, ie, increasing total cholesterol levels, raised concern that it could increase cardiovascular risk. However, total cholesterol levels are increased at the expense of both low-density and high-density lipoprotein particles, in a ratio that may actually confer a protective effect on atherogenesis.Citation101 This, in parallel with the benefit conferred by reducing atherogenic acute-phase proteins (see above), suggests that tocilizumab may be an excellent drug for preventing cardiovascular comorbidity in these patients.

In Europe, tocilizumab is recommended in the 2010 European League Against Rheumatism guidelines only after failure of at least one anti-TNF agent, with or without a synthetic DMARD.Citation42 In the US, tocilizumab is suggested in the 2012 ACR guidelines as an option only after failure of an anti-TNF, abatacept or rituximab,Citation43 although it was approved by the US Food and Drug Administration for moderate to severe RA with inadequate response to any DMARD. Also, in the US the initial recommended dose of tocilizumab is 4 mg/kg while in Europe it is 8 mg/kg. In the large majority of clinical trials, tocilizumab was tested with the 8 mg/kg dose, making this regimen the one most supported by the available evidence. Nevertheless, there have been some reports and registry data supporting the use of the 4 mg/kg dose, especially when there are concerns about toxicity and the patient has achieved remission.Citation102

Beyond tocilizumab

IL-6 signaling has an important singularity: the membrane-bound IL-6 receptor is a hexameric complex composed of a nonsignaling IL-6 binding molecule and two signaling molecules called gp130 (shared by all members of the IL-6 family of receptors).Citation63 Many cell types, upon IL-6 signaling or during apoptosis, shed cleaved soluble molecules of the IL-6 receptor (IL-6R). This soluble version of IL-6R may thereby interact with membrane-bound gp130 in cell types which do not normally express the IL-6R molecule, conferring on them the capacity to be responsive to IL-6 signaling. This process, called trans-signaling,Citation103 is responsible for a fair amount of the recognized actions of IL-6 including its systemic effects, T-cell induction, and chemotaxis. In animal models, blockade of IL-6 trans-signaling with a gp130-Fc fusion protein produced interesting results, alleviating arthritisCitation104 and even reducing atherogenesis. Most importantly, it did better than IL-6R blockade in a sepsis model, conferring 100% survival after cecal puncture.Citation105 The use of trans-signaling-selective blocking agents has not yet been tested in humans, but they could serve as a valid strategy to maintain the major efficacy of tocilizumab with much less toxicity and fewer side effects. Human trials are programmed for 2013, and the results are awaited with some enthusiasm.

On the other hand, inhibition of one of the major intracellular effectors of IL-6 signaling, ie, the JAK-STAT pathway, has been given much attention, and preliminary results are being received with enthusiasm. JAK and STAT molecules have several isoforms which are expressed differently in multiple cell types. They are intracellular messengers for a multitude of kinase cell receptors, including erythropoietin, interferon-gamma, IL-4, and IL-23, apart from IL-6.Citation106 Given its pleiotropic use throughout development, maintenance and regeneration of adult tissue, overt blocking of JAK-STAT signaling would be necessarily fatal, or at least related to serious adverse effects. However, selective inhibition of specific isotypes of JAK or STAT could have a different profile. Inhibition of JAK3 by tofacitinib seems to be almost selective to lymphocytes, reducing its toxicity, and has a major impact on several IL-6 actions (including systemic and cardiovascular effects), inhibiting Th1 and Th17 differentiation and enhancing Treg expansion.Citation107 Phase III trials have already been conducted with an orally available formulation of tofacitinib, which has proved to be successful in reducing clinical and laboratory disease activity and radiographic progression compared with placebo, with a good safety profile. This has been shown at least in methotrexate-inadequate responders (ORAL Scan,Citation108 ORAL Solo,Citation109 and ORAL Standard studies)Citation110 and in other DMARD-inadequate responders (ORAL Sync study),Citation111 and tofacitinib has shown an efficacy and safety profile similar to that of adalimumab, an anti-TNF agent, in association with methotrexate, in the ORAL Standard study. In the ORAL Solo trial, it even demonstrated effectiveness in reaching clinical endpoints in monotherapy.

The forgotten case of anakinra

The IL-1 inhibitor anakinra is a recombinant nonglycosylated form of the human IL-1 receptor antagonist. It has proven to be superior to placebo in RA patients with moderate to severe disease activity in three randomized clinical trials,Citation112Citation114 with differences of up to 42% versus 23% on ACR20 responses (although more modest in ACR50 responses). However, no head-to-head studies have been performed comparing anakinra with other biologics. In a meta-analysis by Thaler et al,Citation115 anakinra seemed to confer no clinical benefit over anti-TNF therapy, and had a worse safety profile, with a greater incidence of infusion reactions (adverse events in 67.2% with anakinra versus 17.5%–22.4% with an anti-TNF). Nevertheless, the data on anakinra are sparse, and for that reason it was not included in the 2012 ACR guidelines for treatment of RA. It is plausible that anakinra could be of interest in specific populations, namely after failure of other biologics, but no studies addressing this have been published.

Conclusion

A fair amount of exciting information has been gathered over the past two decades about the treatment of RA. New biologic agents approved after anti-TNF therapy () have continued to improve the prognosis, quality of life, and life expectancy of RA patients, and a huge number of new agents are being tested in Phase II and III clinical trials, whether targeting new molecules (eg, sarilumab, a monoclonal antibody directed against the alpha subunit of IL-6 receptor, or secukinumab, an anti-IL-17 monoclonal antibody)Citation116 or improving existing drugs (such as ocrelizumab, a humanized version of rituximab).Citation117,Citation118

Table 1 Currently approved molecular targeted strategies to control RA disease activity

Even though great efforts are being undertaken to deepen our understanding of the pathobiology of RA, controversy still exists, and current knowledge fails to explain all the data already gathered. RA is clearly a disease with many subentities, with different outcomes and different potential “optimal therapies”. Synovial tissue histology is a good example, as each different histopathology pattern is correlated with particular treatment responses and clinical characteristics. Subgroup-based studies in RA are still in their infancy, but could provide a means to identify variables predictive of a good biologic treatment response in particular patients.

The outcomes measured in the large majority of clinical trials, and even in basic research, are hitherto centered on arthritis and quality of life scores. Not taking into account all the frequent extra-articular manifestations of RA underestimates the systemic burden of disease, and therefore, it is possible that the results of these trials could be different if a more holistic approach for outcome evaluation had been used. Further, disease activity and bone damage seem to be unlinked during biologic therapy (at least with rituximabCitation119 and tocilizumabCitation120), and this may make the use of current disease activity scores to evaluate the efficacy of such biologic therapies even less adequate. On the other hand, some extra-articular manifestations are often major variables in clinical decision-making (eg, pulmonary disease), and thus should be more extensively studied and considered; present data and guidelines are based on the belief that whatever action improves articular disease will improve other manifestations, which is not necessarily true. Cardiovascular risk, being the major cause of death in this context, has come to attention recently, but the impact of each biologic therapy in relation to others is still understudied.

There is now a wide range of different drugs from different classes of biologic DMARDs available (and soon this number will increase significantly), providing the opportunity to address each patient as a particular case and thereby optimize medical intervention. A strong knowledge of the pharmacologic characteristics of all the drugs, together with a comprehensive approach to each patient, will allow a true informed choice of the best treatment for each case and better overall population-based control of the disease.

Disclosure

The authors report no conflicts of interest in this work.

References

  • MeuneCTouséETrinquartLAllanoreYTrends in cardiovascular mortality in patients with rheumatoid arthritis over 50 years: a systematic review and meta-analysis of cohort studiesRheumatology (Oxford)2009481309131319696061
  • MacGregorAJSniederHRigbyASCharacterizing the quantitative genetic contribution to rheumatoid arthritis using data from twinsArthritis Rheum200043303710643697
  • SilmanAJMacGregorAJThomsonWTwin concordance rates for rheumatoid arthritis: results from a nationwide studyBr J Rheumatol1993329039078402000
  • BoissierMCSemeranoLChallalSSaidenberg-KermanacKNFalgaroneGRheumatoid arthritis: from autoimmunity to synovitis and joint destructionJ Autoimmun20123922222822704962
  • ArvikarSLCollierDSFisherMCClinical correlations with Porphyromonas gingivalis antibody responses in patients with early rheumatoid arthritisArthritis Res Ther201315R10924017968
  • Rantapaa-DahlqvistSde JongBABerglinEAntibodies against cyclic citrullinated peptide and IgA rheumatoid factor predict the development of rheumatoid arthritisArthritis Rheum2003482741274914558078
  • SmolenJLandewéRBMeasePEfficacy and safety of certolizumab pegol plus methotrexate in active rheumatoid arthritis: the RAPID 2 study. A randomised controlled trialAnn Rheum Dis20096879780419015207
  • KeystoneECGenoveseMCKlareskogLGolimumab, a human antibody to tumour necrosis factor {alpha} given by monthly subcutaneous injections, in active rheumatoid arthritis despite methotrexate therapy: the GO-FORWARD StudyAnn Rheum Dis20096878979619066176
  • BuchMHBinghamSJBryerDEmeryPLong-term infliximab treatment in rheumatoid arthritis: subsequent outcome of initial respondersRheumatology (Oxford)2007461153115617478470
  • PaiSPaiLBirkenfeldtRCorrelation of serum IgA rheumatoid factor levels with disease severity in rheumatoid arthritisScand J Rheumatol1998272522569751464
  • van der Helm-van MilAHVerpoortKNBreedveldFCToesREHuizingaTWAntibodies to citrullinated proteins and differences in clinical progression of rheumatoid arthritisArthritis Res Ther20057R949R95816207336
  • EmeryPFleischmannRFilipowicz-SosnowskaAThe efficacy and safety of rituximab in patients with active rheumatoid arthritis despite methotrexate treatment: results of a phase IIB randomized, double-blind, placebo-controlled, dose-ranging trialArthritis Rheum2006541390140016649186
  • TakPPRigbyWFRubbert-RothAInhibition of joint damage and improved clinical outcomes with rituximab plus methotrexate in early active rheumatoid arthritis: the IMAGE trialAnn Rheum Dis201170394620937671
  • MeasePJCohenSGaylisNBEfficacy and safety of retreatment in patients with rheumatoid arthritis with previous inadequate response to tumor necrosis factor inhibitors: results from the SUNRISE trialJ Rheumatol20103791792720194448
  • DassSRawstronACVitalEMHenshawKMcGonagleDEmeryPHighly sensitive B cell analysis predicts response to rituximab therapy in rheumatoid arthritisArthritis Rheum2008582993299918821683
  • VitalEMRawstronACDassSReduced-dose rituximab in rheumatoid arthritisArthritis Rheum20116360360821360489
  • TengYKLevarhtEWToesREHuizingaTWvan LaarJMResidual inflammation after rituximab treatment is associated with sustained synovial plasma cell infiltration and enhanced B cell repopulationAnn Rheum Dis2009681011101618647852
  • SellamJRouanetSHendel-ChavezHBlood memory B cells are disturbed and predict the response to rituximab in patients with rheumatoid arthritisArthritis Rheum2011633692370122127692
  • PisetskyDSGrammerACNingTCLipskyPEAre autoantibodies the targets of B-cell-directed therapy?Nat Rev Rheumatol2011755155621808289
  • LeandroMJCambridgeGEhrensteinMREdwardsJCWReconstitution of peripheral blood B cells after depletion with rituximab in patients with rheumatoid arthritisArthritis Rheum20065461362016447239
  • RollPDörnerTTonyHPAnti-CD20 therapy in patients with rheumatoid arthritis: predictors of response and B cell subset regeneration after repeated treatmentArthritis Rheum2008581566157518512772
  • O’NeillSKShlomchikMJGlantTTCaoYDoodesPDFinneganAAntigen-specific B cells are required as APCs and autoantibody-producing cells for induction of severe autoimmune arthritisJ Immunol20051743781378815749919
  • ChanOTHannumLGHabermanAMMadaioMPShlomchikMJA novel mouse with B cells but lacking serum antibody reveals an antibody-independent role for B cells in murine lupusJ Exp Med19991891639164810330443
  • Tanaka-WatanabeYMatsumotoIIwanamiKB cells play a crucial role as antigen-presenting cells and collaborate with inflammatory cytokines in glucose-6-phosphate isomerase-induced arthritisClin Exp Immunol200915528529419032549
  • TakemuraSKlimiukPABrownAGoronzyJJWeyandCMT cell activation in rheumatoid synovium is B cell dependentJ Immunol20011674710471811591802
  • van de VeerdonkFLLauwerysBMarijnissenRJThe anti-CD20 antibody rituximab reduces the Th17 cell responseArthritis Rheum2011631507151621400475
  • MouraRAGraçaLFonsecaJETo B or not to B: the conducter of rheumatoid arthritis orchestraClin Rev Allergy Immunol20124328129122717935
  • MatsumotoMLoSFCarruthersCJAffinity maturation without germinal centres in lymphotoxin-alpha-deficient miceNature20063824624668684487
  • RennertPDJamesDMackayFBrowningJLHochmanPSLymph node genesis is induced by signaling through the lymphotoxin beta receptorImmunity1998971799697837
  • BuchMHSmolenJSBetteridgeNUpdated consensus statement on the use of rituximab in patients with rheumatoid arthritisAnn Rheum Dis20117090992021378402
  • Martinez-GamboaLBrezinschekHPBurmesterGRDörnerTImmunopathologic role of B lymphocytes in rheumatoid arthritis: rational of B cell-directed therapyAutoimmun Rev2006543744216920569
  • NakouMKatsikasGSidiropoulosPRituximab therapy reduces antivated B cells in both the peripheral blood and bone marrow of patients with rheumatoid arthritis: depletion of memory B cells correlates with clinical responseArthritis Res Ther200911R13119715572
  • KavanaughARosengrenSLeeSJAssessment of rituximab’s immunomodulatory synovial effects (ARISE trial). 1: clinical and synovial biomarker resultsAnn Rheum Dis20086740240817644541
  • GheitaTAGheitaHAKenawySARituximab restored the muscle power and rescued from a refractory fatal respiratory failure in a patient with elderly-onset polymyositisJoint Bone Spine20127910110221840235
  • Rubbert-RothATakPPZerbiniCEfficacy and safety of various repeat treatment dosing regimens of rituximab in patients with active rheumatoid arthritis: results of a phase III randomized study (MIRROR)Rheumatology (Oxford)2010491683169320463186
  • CohenSBEmeryPGreenwaldMWRituximab for rheumatoid arthritis refractory to anti-tumor necrosis factor therapy: results of a multicenter, randomized, double-blind, placebo-controlled, phase III trial evaluating primary efficacy and safety at twenty-four weeksArthritis Rheum2006542793280616947627
  • KeystoneECCohenSBEmeryPMultiple courses of rituximab produce sustained clinical and radiographic efficacy and safety in patients with rheumatoid arthritis and an inadequate response to 1 or more tumor necrosis factor inhibitors: 5-year data from the REFLEX studyJ Rheumatol2012392238224623027887
  • Gomez-ReinoJJManeiroJRRuizJComparative effectiveness of switching to alternative tumour necrosis factor (TNF) antagonists versus switching to rituximab in patients with rheumatoid arthritis who failed previous TNF antagonists: the MIRAR StudyAnn Rheum Dis2012711861186422736086
  • SolimanMMHyrichKLLuntMWatsonKDSymmonsDPAsh-croftDMRituximab or a second anti-tumor necrosis factor therapy for rheumatoid arthritis patients who have failed their first anti-tumor necrosis factor therapy? Comparative analysis from the British Society for Rheumatology Biologics RegisterArthritis Care Res20126411081115
  • EmeryPDeodharARigbyWFEfficacy and safety of different doses and retreatment of rituximab: a randomised, placebo-controlled trial in patients who are biological naive with active rheumatoid arthritis and an inadequate response to methotrexate (Study Evaluating Rituximab’s Efficacy in MTX iNadequate rEsponders (SERENE)Ann Rheum Dis2010691629163520488885
  • ChatzidionysiouKLieENasonovEEffectiveness of disease-modifying antirheumatic drug co-therapy with methotrexate and leflunomide in rituximab-treated rheumatoid arthritis patients: results of a 1-year follow-up study from the CERERRA collaborationAnn Rheum Dis20127137437721972242
  • SmolenJSLandewéRBreedveldFCEULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatid drugsAnn Rheum Dis20106996497520444750
  • SinghJAFurstDEBharatA2012 Update of the 2008 American College of Rheumatology recommendations for the use of diseas-modifying antirheumatic drugs and biologic agents in the treatment of theumatoid arthritisArthritis Care Res (Hoboken)20126462563922473917
  • CantaertTKollnJTimmerTB lymphocyte autoimmunity in rheumatoid synovitis is independent of ectopic lymphoid neogenesisJ Immunol200818178579418566445
  • NevesMAlvesJDFactors implicated in the generation and persistence of long-lived plasma cell-mediated autoimmunityAutoimmun Rev20111037538221224017
  • HumbyFBombardieriMManzoAEctopic lymphoid structures support ongoing production of class-switch autoantibodies in rheumatoid synoviumPLoS Med200965975
  • KlimiukPAGoronzyJJBjörnssonJBeckenbaughRDWeyandCMTissue cytokine patterns distinguishes variants of rheumatoid synovitisAm J Pathol1997151131113199358757
  • FonsecaJECanhãoHResendeCHistology of the synovium tissue: value of semi-quantitative analysis for the prediction of joint erosions in rheumatoid arthritisClin Exp Rheumatol20001855956411072594
  • MeiHESchmidtSDörnerTRational of anti-CD19 immunotherapy: an option to target autoreactive plasma cells in autoimmunityArthritis Res Ther201214Suppl 5S123281743
  • YazawaNHamaguchiYPoeJCTedderTFImmunotherapy using unconjugated CD19 monoclonal antibodies in animal models for B lymphocyte malignancies and autoimmune diseaseProc Natl Acad Sci U S A2005102151781518316217038
  • HamaguchiYUchidaJCainDWThe peritoneal cavity provides a protective niche for B1 and conventional B lymphocytes during anti-CD20 immunotherapy in miceJ Immunol20051744389439915778404
  • TedderTFCD19: a promising B cell target for rheumatoid arthritisNat Rev Rheumatol2009557257719798033
  • StoneMJSausvilleEAFayJWA Phase I study of bolus versus continuous infusion of the anti-CD19 immunotoxin, IgG-HD37-dgA, in patients with B-cell lymphomaBlood199688118811978695836
  • YounesAKimSRomagueraJPhase I multidose-escalation study of the anti-CD19 maytansinoid immunoconjugate SAR3419 administered by intravenous infusion every 3 weeks to patients with relapsed/refractory B-cell lymphomaJ Clin Oncol2012302776278222753910
  • JinXDingCBelimumab – an anti-BLyS human monoclonal antibody for rheumatoid arthritisExpert Opin Biol Ther20131331532223268610
  • GenoveseMCKinnmanNde La BourdonnayeGPena RossiCTakPPAtacicept in patients with rheumatoid arthritis and an inadequate response to tumor necrosis factor antagonist therapy: results of a Phase II, randomized, placebo-controlled, dose-finding trialArthritis Rheum2011631793180321452293
  • StohlWMerrillJTMcKayJDEfficacy and safety of belimumab in patients with rheumatoid arthritis: a Phase II, randomized, double-blind, placebo-controlled, dose-ranging studyJ Rheumatol20134057958923547209
  • van VollenhovenRFKinnmanNVincentEWaxSBathonJAtacicept in patients with rheumatoid arthritis and an inadequate response to methotrexateArthritis Rheum2011631782179221452294
  • DillonSRHarderBLewisKBB-lymphocyte stimulator/a proliferation-inducing ligand heterotrimers are elevated in the sera of patients with autoimmune disease and are neutralized by atacicept and B-cell maturation antigen-immunoglobulinArthritis Res Ther201012R4820302641
  • FiresteinGSEtiology and pathogenesis of rheumatoid arthritisFiresteinGSBuddRCHarrisEDJMcInnesIBRuddySSergentJSKelley’s Textbook of Rheumatology8th edPhiladelphia, PASaunders2009
  • KoendersMILubbertsEvan de LooFAJInterleukin-17 acts independently of TNF-alpha under arthritic conditionsJ Immunol20061766262626916670337
  • DienzORinconMThe effects of IL-6 on CD4 T cell responsesClin Immunol2009130273318845487
  • KishimotoTIL-6: from its discovery to clinical applicationsInt Immunol20102234735220410258
  • SzodorayPNakkenBBarathSAltered Th17 cells and Th17/regulatory T-cell ratios indicate the subsequent conversion from undifferentiated connective tissue disease to definitive systemic autoimmune disordersHum Immunol8202013 [Epub ahead of print.]
  • MatsukiFSaegusaJMiyamotoYMisakiKKumagaiSMorinobuACD45RA-Foxp3high activated/effector regulatory T cells in the CCR7+CD45RA-CD27+CD28+ central memory subset are the decreased in peripheral blood from patients with rheumatoid arthritisBiochem Biophys Res Commun201343877878323747721
  • EsenstenJHWofsyDBluestoneJARegulatory T cells as therapeutic targets in rheumatoid arthritisNat Rev Rheumatol2009556056519798031
  • CeerazSHallCChoyESpencerJCorrigallVMDefective CD8+CD28-regulatory T cell suppressor function in rheumatoid arthritis is restored by TNF inihibitor therapyClin Exp Immunol2013174182623786396
  • SzalayBVásárhelyiBCsehAThe impact of conventional DMARD and biological therapies on CD4+ cell subsets in rheumatoid arthritis: a follow-up studyClin Rheumatol8112013 [Epub ahead of print.]
  • SolomonGET-cell agents in the treatment of rheumatoid arthritisBull NYU Hosp Jt Dis20127019119423259627
  • BuchMHBoyleDLRosengrenSMode of action of abatacept in rheumatoid arthritis patients having failed tumour necrosis factor blockade: a histological, gene expression and dynamic magnetic resonance imaging pilot studyAnn Rheum Dis2009681220122718772191
  • WeismanMHDurezPHalleguaDReduction of inflammatory biomarker response by abatacept in treatment of rheumatoid arthritisJ Rheumatol2006332162216617014006
  • von KempisJDudlerJHaslerPUse of abatacept in rheumatoid arthritisSwiss Med Wkly2012142w1358122581564
  • GottenbergJERavaudPCantagrelAPositivity for anti-cyclic citrullinated peptide is associated with a better response to abatacept: data from the ‘Orencia and Rheumatoid Arthritis’ registryAnn Rheum Dis2012711815181922615458
  • KremerJMGenantHKMorelandLWEffects of abatacept in patients with methotrexate-resistant active rheumatoid arthritis: a randomized trialAnn Intern Med200614486587616785475
  • SchiffMKeisermanMCoddingCEfficacy and safety of abatacept or infliximab vs placebo in ATTEST: a phase III, multi-centre, randomised, doubleblind, placebo-controlled study in patients with rheumatoid arthritis and an inadequate response to methotrexateAnn Rheum Dis2008671096110318055472
  • GenoveseMCBeckerJCSchiffMAbatacept for rheumatoid arthritis refractory to tumor necrosis factor alpha inhibitionN Engl J Med20053531114112316162882
  • SchiffMKeisermanMCoddingCClinical response and tolerability to abatacept in patients with rheumatoid arthritis previously treated with infliximab or abatacept: open-label extension of the ATTEST StudyAnn Rheum Dis2011702003200721914628
  • SchiffMPritchardCHuffstutterJEThe 6-month safety and efficacy of abatacept in patients with rheumatoid arthritis who underwent a washout after anti-tumour necrosis factor therapy or were directly switched to abatacept: the ARRIVE trialAnn Rheum Dis2009681708171419074911
  • WesthovensRRoblesMXimenesACClinical efficacy and safety of abatacept in methotrexate naive patients with early rheumatoid arthritis and poor prognostic factorsAnn Rheum Dis2009681870187719124524
  • SchiffMSubcutaneous abatacept for the treatment of rheumatoid arthritisRheumatology (Oxford)20135298699723463804
  • NashPNayiagerSGenoveseMCImmunogenicity, safety, and efficacy of abatacept administered subcutaneously with or without background methotrexate in patients with rheumatoid arthritis: results from a phase III, international, multicenter, parallel-arm, open-label studyArthritis Care Res (Hoboken)20136571872823097311
  • PieperJHerrathJRaghavanSMuhammadKvan VollenhovenRMalmströmVCTLA4-Ig (abatacept) therapy modulates T cell effector functions in autoantibody-positive rheumatoid arthritis patientsBMC Immunol2013143423915385
  • WeinblattMSchiffMGoldmanASelective co-stimulation modulation using abatacept in patients with active rheumatoid arthritis while receiving etanercept: a randomized clinical trialAnn Rheum Dis20076622823416935912
  • BrennenFMMcLnnesIVEvidence that cytokines play a role in rheumatoid arthritisJ Clin Invest20081183537354518982160
  • ChoyEUnderstanding the dynamics: pathways involved in the pathogenesis of rheumatoid arthritisRheumatology (Oxford)201251Suppl 5v3v1122718924
  • GerneroPThompsonEWoodworthTSmolenJSRapid 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 methotrexateArthritis Rheum201062334320039425
  • DayerJMChoyETherapeutic targets in rheumatoid arthritis: the interleukin-6 receptorRheumatology (Oxford)201049152419854855
  • NunomuraWHatakeyamaM[Binding of low density lipoprotein (LDL) to C-reactive protein (CRP): a possible binding through apolipo-protein B in LDL at phosphorylcholine-binding site of CRP]Hokkaido Igaku Zasshi199065474480 Japanese2227796
  • MattssonNMagnussenCGRönnemaaTMetabolic syndrome and carotid intima-media thickness in young adults: roles of apolipoprotein B, apolipoprotein A-I, C-reactive protein, and secretory phospholipase A2: the cardiovascular risk in young Finns studyArterioscler Thromb Vasc Biol2010301861186620539018
  • ArtlAMarscheGLestavelSSattlerWMalleERole of serum amyloid A during metabolism of acute-phase HDL by macrophagesArterioscler Thromb Vasc Biol20002076377210712402
  • BankaCLYuanTde BeerMCKindyMCurtissLKde BeerFCSerum amyloid A (SAA): influence on HDL-mediated cellular cholesterol effluxJ Lipid Res199536105810657658153
  • LeeHYKimSDBaekSHSerum amyloid A stimulates macrophage foam cell formation via lectin-like oxidized low-density lipo-protein receptor 1 upregulationBiochem Biophys Res Commun2013433182323454129
  • PatelAMMorelandLWInterleukin-6 inhibition for treatment of rheumatoid arthritis: a review of tocilizumab therapyDrug Des Dev Ther20104263278
  • SmolenJSBeaulieuARubbert-RothAEffect of interleukin-6 receptor inhibition with tocilizumab in patients with rheumatoid arthritis (OPTION study): a double-blind, placebo-controlled, randomized trialLancet200837198799718358926
  • FleischmannRMHallandAMBrzoskoMTocilizumab inhibits structural joint damage and improves physical function in patients with rheumatoid arthritis and inadequate responses to methotrexate: LITHE study 2-year resultsJ Rheumatol201340211312623322466
  • GenoveseMCMcKayJDNasonovELInterleukin-6 receptor inhibition with tocilizumab reduces disease activity in rheumatoid arthritis with inadequate response to diseasemodifying antirheumatic drugs: the tocilizumab in combination with traditional disease-modifying antirheu-matic drug therapy studyArthritis Rheum200858102968298018821691
  • GenoveseMCMcKayJDNasonovELInterleukin-6 receptor inhibition with tocilizumab reduces disease activity in rheumatoid arthritis with inadequate response to diseasemodifying antirheumatic drugs: the tocilizumab in combination with traditional disease-modifying antirheumatic drug therapy studyArthritis Rheum200858102968298018821691
  • DougadosMKisselKSheeranTAdding 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 Dis201372435022562983
  • JonesGSebbaAGuJComparison of tocilizumab mono-therapy versus methotrexate monotherapy in patients with moderate to severe rheumatoid arthritis: the AMBITION studyAnn Rheum Dis2010691889619297346
  • SalliotCvan der HeijdeDLong-term safety of methotrexate monotherapy in patients with rheumatoid arthritis: a systematic literature researchAnn Rheum Dis2009681100110419060002
  • HanselBBruckertE[Lipid profile and cardiovascular risk in patients with rheumatoid arthritis: effect of the disease and of drug therapy]Ann Endocrinol (Paris)201071257263 French20416859
  • OliveiraSPatient with a 9-year history of rheumatoid arthritis and hepatitis B co-infection who responds favorably to tocilizumab mono-therapy after progression or intolerance to several disease-modifying anti-rheumatic drugsAdv Ther201229 Special Issue1213
  • Rose-JohnSIL-6 Trans-signaling via the soluble IL-6 receptor: importance for the pro-inflammatory activities of IL-6Int J Biol Sci201281237124723136552
  • NowellMAWilliamsASCartySATherapeutic targeting of IL-6 trans signaling counteracts STAT3 control of experimental inflammatory arthritisJ Immunol200918261362219109195
  • BarkhausenTTschernigTRosenstielPSelective blockade of interleukin-6 trans-signaling improves survival in a murine polymicrobial sepsis modelCrit Care Med2011391407141321336117
  • MurrayPJThe JAK-STAT signaling pathway: input and output integrationJ Immunol20071782623262917312100
  • TanakaYYamaokaKJAK inhibitor tofacitinib for treating rheumatoid arthritis: from basic to clinicalMod Rheumatol20132341542423212593
  • van der HeijdeDTanakaYFleischmannRTofacitinib (CP-690,550) in patients with rheumatoid arthritis receiving methotrexate: twelve-month data from a twenty-four-month phase III randomized radiographic studyArthritis Rheum201365355957023348607
  • FleischmannRKremerJCushJPlacebo-controlled trial of tofacitinib monotherapy in rheumatoid arthritisN Eng J Med20123676495507
  • van VollenhovenRFFleischmannRCohenSTofacitinib or adalimumab versus placebo in rheumatoid arthritisN Eng J Med20123676508519
  • KremerJLiZGHallSTofacitinib in combination with nonbiologic disease-modifying antirheumatic drugs in patients with active rheumatoid arthritis: a randomized trialAnn Intern Med2013159425326124026258
  • BresnihanBAlvaro-GraciaJMCobbyMTreatment of rheumatoid arthritis with recombinant human interleukin-1 receptor antagonistArthritis Rheum199841219622049870876
  • CohenSHurdECushJTreatment of rheumatoid arthritis with anakinra, a recombinant human interleukin-1 receptor antagonist, in combination with methotrexate: results of a twenty-four-week, multicenter, randomized, double-blind, placebo-controlled trialArthritis Rheum20024661462411920396
  • CohenSBMorelandLWCushJJA multicentre, double blind, randomised, placebo controlled trial of anakinra (Kineret), a recombinant interleukin 1 receptor antagonist, in patients with rheumatoid arthritis treated with background methotrexateAnn Rheum Dis2004631062106815082469
  • ThalerKChandiramaniDVHansenRAGeraldGEfficacy and safety of anakinra for the treatment of rheumatoid arthritis: an update of the Oregon Drug Effectiveness Review ProjectBiologics2009348549820054439
  • ReichertJMWhich are the antibodies to watch in 2013?MAbs201351423254906
  • TakPPMeasePJGenoveseMCSafety and efficacy of ocrelizumab in patients with rheumatoid arthritis and an inadequate response to at least one tumor necrosis factor inhibitor: results of a forty-eight-week randomized, double-blind, placebo-controlled, parallel-group phase III trialArthritis Rheum20126436037022389919
  • RigbyWTonyHPOelkeKSafety and efficacy of ocrelizumab in patients with rheumatoid arthritis and an inadequate response to methotrexate – results of a forty-eight-week randomized, double-blind, placebo-controlled, parallel-group Phase III trialArthritis Rheum20126435035921905001
  • AletahaDAlastiFSmolenJSRituximab dissociates the tight link between disease activity and joint damage in rheumatoid arthritis patientsAnn Rheum Dis20137271222915619
  • SmolenJSMartinez AvilaJCAletahaDTocilizumab inhibits progression of joint damage in rheumatoid arthritis irrespective of its anti-inflammatory effects: dissociation of the link between inflammation and destructionAnn Rheum Dis20127168769322121130