78
Views
4
CrossRef citations to date
0
Altmetric
Original Research

Personalization of biologic therapy in patients with rheumatoid arthritis: less frequently accounted choice-driving variables

, , , , , , , , , , & show all
Pages 2097-2111 | Published online: 24 Oct 2018

Abstract

Objective

To propose appropriate statements that drive the choice of biologic therapies in patients with rheumatoid arthritis (RA), factoring in their impact on the following issues: anti-drug antibody (ADAb) formation, suspicion and management of infections, lupus-like syndrome (LLS), effects on bone mass and sexual sphere, and relationship between RA and periodontal disease (PD).

Methods

An overview of existing evidence was undertaken by an expert panel on behalf of the Italian board for the TAilored BIOlogic therapy (ITABIO). Data were extracted from controlled trials, national registries, national health care databases, post-marketing surveys, and, when required by the paucity of controlled studies, from open-label clinical series. Anti-tumor necrosis factor (anti-TNF) and non-anti-TNF-targeted biologics approved for RA were investigated.

Results

ADAb formation is chiefly associated with anti-TNFs, and it is reduced by combination therapy with methotrexate. To date, ADAb titration is not advisable for clinical practice, and, in case of anti-TNF secondary failure, a non-anti-TNF biologic is indicated. LLS is observed in anti-TNF receivers and, in most cases, resolves without anti-TNF withdrawal. A non-anti-TNF biologic is advisable in patients experiencing LLS. Non-anti-TNFs demonstrated a low or absent infection risk and are preferable in patients with comorbidities. Due to their positive effects on bone mass, anti-TNFs are indicated in women at osteoporosis risk, whereas non-anti-TNF have been poorly investigated. The emerging evidence of the relationship between RA and PD and the effects on anti-TNF efficacy should lead clinicians to consider the periodontal status in RA patients. Anti-TNFs may exert a positive effect on fertility and sexuality, and clinicians should explore these aspects in RA patients.

Conclusion

The optimization of biologic therapies by taking into proper account the above issues would improve patient outcomes.

Introduction

The results of randomized clinical trials (RCTs) and real-life reports, with special reference to national registries, have clarified many of the aspects of biologic disease-modifying anti-rheumatic drugs (bDMARD) in patients with inflammatory rheumatic disorders, including the mechanism of action, efficacy and safety, availability of response predictors, and possibility of optimizing their use in single patients.Citation1 However, some biologic-related issues that may influence therapeutic choice are often disregarded, or still need to be fully addressed, by clinicians; these include the immunogenicity, suspicion and management of infections, anti-tumor necrosis factor (TNF)-related lupus-like syndrome (LLS), skeletal effects of biologics, relationship between periodontal disease (PD) and rheumatoid arthritis (RA), and effects of biologics on both fertility and sexuality. Indeed, rheumatologists are very careful in evaluating the disease activity and clinical course of RA, but an overshadowing of several features for a complete patient management frequently occurs. By contrast, the abovementioned topics may play a pivotal role for the best outcome, not only in terms of articular response but also for the improvement of the overall quality of care and the prevention of complications.

With these considerations in mind, the present article was intended to formulate practical indications on the impact of immunogenicity, infections, and LLS; effects of biologics on the skeleton; role of coexisting PD in patients with RA; and influence of RA and related therapies on fertility and sexuality.

Methods

As previously reported,Citation1 a multidisciplinary expert panel on behalf of the Italian board for the TAilored BIOlogic therapy (ITABIO) reviewed the literature on the existing evidence on the immunogenicity of biologics, suspicion and management of infections, LLS, biologic skeletal effects, association of RA with PD and its impact on both disorders and on biologic therapy, impairment of fertility and sexual sphere, and effects of biologics. Taking in account the emerging evidence on these topics, appropriate statements useful to therapy tailoring were formulated.

Literature search

The literature review was conducted by using the PubMed database to identify English-language articles related to the abovementioned topics. Data were extracted from RCTs, national registries of biologics, national health care databases, and post-marketing surveys. When these source data were not available for specific topics, the evidence was retrieved from open-label studies on variable sample-size clinical series.

The following drugs were investigated: infliximab (IFX), etanercept (ETN), adalimumab (ADA), golimumab (GOL), certolizumab pegol (CZP), tocilizumab (TCZ), abatacept (ABA), and rituximab (RTX). The research was conducted by crossing each drug name with the following key terms: RA, early RA (ERA), efficacy, safety, immunogenicity, anti-drug antibody, infection, opportunistic infection, tuberculosis, osteoporosis, bone mass, skeletal manifestations, periodontal disease, periodontitis, fertility, and sexuality. The literature review was extended to literature published up to April 30, 2018.

Mechanism of immunogenicity and fallout on clinical practice

It has long been recognized that biologics used for treatment of RA can give rise to the development of antibodies targeting the respective biodrug, designated as “antidrug antibodies” (ADAbs).Citation2Citation4 ADAb induction is commonly a polyclonal response, resulting in the formation of antibodies that may differ widely in terms of serum titer, antigenic affinity, and biologic actions. Accordingly, immunogenic responses against biologics give rise to pools of antibodies that comprise different subtypes (commonly, immunoglobulin M [IgM] and IgG, but, in a minority of cases, IgE as well) and isotypes (eg, IgG1, IgG2, IgG3), targeting different epitopes of the same biodrug and displaying different binding patterns.Citation2 Based on the targeted epitopes and their intrinsic biological properties, ADAbs are currently classified as non-neutralizing and neutralizing antibodies. The former, usually comprising IgM, IgG, and IgE, bind epitopes located in molecular domains of the biodrug that are not critical for the interaction with its molecular target, which usually accelerates the biodrug clearance (clearing antibodies), enhances the bioavailability (sustaining antibodies), or elicits hypersensitivity adverse reactions.Citation5,Citation6

By contrast, neutralizing antibodies (usually IgG) bind epitopes that are critical for the interaction of the biodrug with its molecular target, thereby impairing its therapeutic efficacy, or eliciting adverse reactions through suppression of the biological actions of an endogenous mediator. Furthermore, similarly as non-neutralizing ADAbs, neutralizing ADAbs may affect the biodrug pharmacokinetic profile, mainly preventing its absorption from the injection site, and/or can elicit hypersensitivity reactions. In addition, as compared to neutralizing ADAbs, non-neutralizing ADAbs are usually characterized by higher titer, early induction, and prolonged persistence throughout the treatment period.Citation5,Citation6

The immune-pathogenesis of ADAbs relies basically on two main mechanisms. Most commonly, the biodrug is processed by antigen-presenting cells with subsequent presentation of its antigenic moieties to competent T lymphocytes, which undergo activation and in turn activate B lymphocytes, followed by clonal expansion and differentiation into plasma cells, which initiate ADAb production. The second mechanism, which is T-cell-independent, occurs usually when a number of biodrug molecules cluster to form macromolecular aggregates that behave as polyvalent antigenic structures. Such aggregates have the potential of promoting B-cell-receptor clustering on the membrane surface of B lymphocytes, thus stimulating their direct activation, which is followed by clonal expansion, differentiation into plasma cells, and initiation of ADAb production.Citation7 In some instances, the immunogenic response triggered by the biodrug amplifies (epitope spreading) with a first phase of ADAb production through the abovementioned clustering mechanism and a second phase where ADAb–biodrug complexes are taken up by naïve B lymphocytes which then behave as antigen-presenting cells to process the biodrug and present its antigenic moieties to competent T lymphocytes; this results in the production of more potent ADAbs targeted against different epitopes than those that were involved in the first phase.Citation5,Citation8

In patients developing ADAbs by the mechanism of direct B-cell activation, the possible presence of biologic impurities – in combination with the disease-related inflammatory background – can behave as danger signals, leading to the stimulation of toll-like receptors (TLRs); this is followed by a further amplification of activated B lymphocytes and results in a switch from low-affinity IgM to high-affinity IgG ADAbs, with a consequent increased risk of detrimental clinical consequences.Citation5,Citation9

ADAb titration

Several attempts have been made to set up assay methods suitable for testing both the presence and titer of ADAbs as well as to follow-up their variations over the time in the sera of patients. Ideally, to fulfill the purpose of generating clinically useful results, laboratory methods for the assessment of ADAbs should be suitable for selectively detecting antibodies of any isotype that may develop against the different epitopes of the biodrug, and they should be sensitive enough for detecting both low- and high-affinity ADAbs.Citation4 Unfortunately, ADAb titration assays have yielded highly variable results to such an extent that the actual significance of ADAb testing in clinical practice remains a matter of debate. This extreme variability depends largely on severe methodological limitations affecting the available assays, as extensively discussed in previous reviews.Citation3,Citation4,Citation10 Thus, given the unique technical features and peculiar drawbacks of each assay method, comparative analyses of immunogenicity results obtained by means of different methods are inappropriate, likely to lead to biased conclusions, and should therefore be avoided.Citation4 This statement is reinforced by the wide percentages of ADAb positivity associated with each biodrug, ranging from 0% to 83% for IFX, 0% to 54% for ADA, 3% to 37% for CZP, 0% to 19% for GLM, and 0% to 13% for ETN.Citation11

At least five additional major factors can account for the heterogeneity of results on ADAb assessments, and these include: 1) sampling procedures (sampling time and sample handling); 2) patients’ characteristics in terms of intrinsic propensity to develop immunogenic responses; RA disease activity and/or duration; and presence of comorbidities; 3) intrinsic biodrug molecular properties as well as product composition and formulation, including different immunogenic molecular domains, presence of murine moieties, different glycosylation patterns, and presence of impurities or aggregates in the formulation; 4) differences in administration routes and treatment regimens, with greater immunogenicity promotion by subcutaneous administration and intermittent treatment; and 5) combination therapy with methotrexate (MTX), although not completely explained, reduces the immunogenicity of anti-TNFs.Citation2,Citation12Citation14 However, in the real-life world, approximately 30% of RA patients receive anti-TNF monotherapy,Citation1 and, at least in part, the lower retention rate of these biologics, as observed with such a therapeutic regimen, is related to ADAbs.

Immunogenicity of individual biologics

Focusing on the immunogenicity of anti-TNFs, ADAbs can target their Fab or Fc fragment. With specific regard for IFX, ADA, GOL, and CZP, the majority of ADAbs target their Fab fragment and are endowed with neutralizing activity. Such a pattern of immunogenic response likely results from antiidiotype reactions, and this may explain why the development of humanized therapeutic antibodies has decreased – although not suppressed – the risk of ADAb induction. ADAbs targeting the Fc fragment (particularly, the hinge region) do not exert neutralizing activity, but can significantly decrease their circulating levels by enhancing the biodrug pharmacokinetic clearance. On the other hand, receptor fusion proteins, such as ETN or ABA, can promote the induction of ADAbs directed against the receptor moiety (mainly exerting neutralizing activity) or the antibody Fc fragment (mainly, the hinge region). However, current evidence on ETN suggests only the development of non-neutralizing ADAbs.Citation6,Citation13,Citation14

According to three systematic reviews,Citation14Citation16 the majority of clinical studies agree on at least three points of evidence: serum ADAb positivity appears to be a risk factor for a loss of therapeutic efficacy; the development of ADAbs increases the risk of immune adverse reactions, which include infusion reactions, injection-site reactions, and, more occasionally, serum sickness and Arthus reactions; and concomitant treatment with MTX or azathioprine appears to decrease the risk of ADAb formation against anti-TNFs. Several authors have indeed reported that ADAbs can be responsible for decrease in biodrug concentration, leading to secondary treatment failure,Citation14Citation16 and similar data have been yielded by real-world clinical practice non-interventional studies.Citation17

With regard to non-anti-TNF-targeted biologics, the risk of ADAb development seems consistently reduced. In a recent review, ADAb positivity was recorded in 1.5% of RA patients receiving intravenous or subcutaneous TCZ, with no differences between TCZ monotherapy or combination therapy with conventional synthetic DMARDs.Citation18 Similar results have been achieved in studies on RA patients that were designed specifically to correlate serum ADAb titers and TCZ levels with disease activity.Citation19,Citation20 Likewise, low ADAb positivity percentages have been reported with ABA (2%–20%) and RTX (0%–21%).Citation11

Infection suspicion and diagnosis in patients taking biologics

RA itself is associated with an increased in the risk of infections,Citation21 and the risk is increased in patients with severe disease requiring biologics, especially anti-TNFs.Citation22

Anti-TNFs increase the risk of infection by different mechanisms: 1) impairment of the maintenance of granulomas,Citation23,Citation24 leading to increased risk of new tuberculosis infection, reactivation of latent tuberculosis,Citation25 and other granulomatous infections, such as Histoplasma capsulatumCitation26 and 2) impairment of phagosome formation and clearance of intracellular pathogens;Citation24 biologic-induced neutropenia predisposing to infections such as Candida or Aspergillus,Citation27 or interference with immune response against viral infections and consequent enhanced complications in patients infected with hepatitis B virus (HBV)Citation28 or varicella zoster virus (VZV).Citation29,Citation30

However, more frequently, the serious infections associated with biologics are bloodstream infections, septic arthritis, osteomyelitis, pneumonia, urinary tract infections with urosepsis and pyelonephritis, soft-tissue cellulitis, gastroenteritis, and intra-abdominal infections. The risk of serious infections is highest in the first 6 months of biologic treatment, plateauing off by 24–36 months.Citation31 The results of most analyses show that, among anti-TNFs, IFX is associated with a higher risk of serious infections than ETN. Concerning the non-anti-TNF-targeted biologics, the results of the head-to-head RCT of ADA and ABA (AMPLE study) showed a comparable occurrence of infections in the two treatment arms.Citation32 However, it should be noted that patients treated in real-world practice differ from those, highly selected subjects enrolled in RCTs,Citation33,Citation34 and data from registries and observational studies demonstrated a low or absent infection risk in patients receiving non-anti-TNF-targeted biologics, including TCZ, ABA, and RTX.Citation35

Clinical presentation and diagnostic approach to severe infections

reports the clinical presentation and the major diagnostic approaches useful to diagnose severe infections in patients treated with biologics. Herein, we describe mainly the bloodstream infection, which is a life-threatening condition that arises when the body’s response to infection injures its own tissues.Citation1,Citation36Citation41 There may be symptoms related to a specific infection, such as a cough with pneumonia or painful urination with a kidney infection. In people with a weakened immune system and in the very young or old people, there may be no symptoms of a specific infection and the body temperature may be low or normal. Severe sepsis causes poor organ function or insufficient blood flow, leading to low blood pressure, high blood lactate, or low urine output and, finally, septic shock.

Table 1 Clinical presentation and diagnostic procedures of the most common serious infections associated with the use of biologics

Furthermore, a small but significant risk of serious opportunistic infection is associated with anti-TNF-α therapies.Citation30 Opportunistic infection is defined as a usually serious and progressive infection by an organism that, under normal circumstances, possesses little or no pathologic capabilities. However, predisposing factors such as underlying disease or medical treatment can reduce the patient’s immune response, permitting the organism to cause an infectious disease.Citation21 Beyond the role of biologics, in patients with RA, several adjunctive risks may lead to decreased innate immune function and increased risk of opportunistic infections, such as aging, diabetes, alcoholism, concomitant immunosuppressive therapies, malnutrition, and micronutrient deficiency.Citation21,Citation30,Citation39,Citation41

Among those considered as opportunistic infections, we should consider bacterial, fungal, viral, and parasitic infections.Citation30 When possible, patients should be adequately vaccinated and closely monitored for early signs of infection.Citation42

Anti-TNF and LLS

Since the introduction of anti-TNF therapy, there have been reports on autoantibody production and of lupus-like manifestations associated with these therapies.Citation43Citation51

The appearance of antinuclear antibodies (ANA) during anti-TNF treatment has been described in the initial IFX trials and also confirmed after exposure to other anti-TNF drugs such as ETN and ADA.Citation43Citation45 The development of ANA or increase of ANA titer in previously positive patients when on anti-TNFs has been reported in percentages ranging from 20% up to 70%. In addition, the appearance of anti-double-strand DNA (anti-dsDNA) antibodies and anticardiolipin antibodies (usually IgM) has been reported.

Drug-induced lupus erythematosus (DILE) has long been recognized as a condition associated with a number of different medications,Citation45 but anti-TNF-induced-LLS has specific clinical and serological characteristics and differs from the classical DILE in many aspects.Citation46Citation49

The true incidence of LLS is difficult to be established but it appears low, being reported in 0.18%–0.41% of patients treated with different anti-TNFs.Citation46Citation49 The mean latency until the first manifestation is in the range from 2 to 40 weeks and the female:male ratio is 5:1. LLS has been reported in anti-TNF-treated patients with RA, psoriatic arthritis, and inflammatory bowel disease.Citation46

The clinical picture of LLS is characterized by fever and constitutional symptoms (70%), high frequency of cutaneous manifestations (malar rash, discoid rash in up to 70%), arthritis (50%), myalgias (50%), and serositis (18%), whereas the renal system and CNS are affected more rarely (<10% of patients).Citation48,Citation49 Serologically, LLS is characterized by the positivity of anti-dsDNA antibodies and lower positivity of anti-histone antibodies (50%) and hypo-complementemia (60%).Citation47Citation51

In the majority of cases, LLS resolves with anti-TNF withdrawal; however, up to 40% of patients require corticosteroids and a smaller percentage (approximately 10%) require immunosuppressive therapy.Citation47

Potential mechanisms for LLS development

TNF plays a double role in systemic lupus erythematosus (SLE). Some studies, in fact, have shown an increased expression of TNF in kidney biopsies and in skin of patients with SLE, thus suggesting a possible pathogenic role of this cytokine.

Indeed, there are data in the literature showing a positive effect of short-duration anti-TNF therapy in the treatment of class V glomerulonephritis and joint involvement in SLE. No major side effects have been reported and there were no disease flares, although a transient increase of anti-dsDNA antibodies titers and anticardiolipin antibodies has been described in these patients during therapy.Citation52,Citation53 However, in animal models, a reduction of TNF predisposes to autoimmunity and antibody development. This might be due to the suppression of Th1 cytokines and a subsequent increase of Th2 cytokines, leading to increased autoantibodies production. Alternatively, the reduction of TNF may interfere with apoptosis thereby leading to a reduced clearance of nuclear antigens, consequently leading again to increased antibody production.Citation54

Is ANA assessment to be recommended prior to anti-TNF therapy initiation?

No data in the literature thus far suggest that ANA positivity prior to therapy initiation has a prognostic role for LLS development, and there are no data supporting the need for antibody testing at baseline.Citation54,Citation55 However, it has been hypothesized that patients developing LLS may have a latent or subclinical form of SLE and, therefore, a careful history aimed at assessing the clinical manifestations is suggested.

Biologic agents and skeletal effects in RA

Systemic osteoporosis (OP) and increased fracture rates are well-known comorbidities in RA.Citation56,Citation57 Improved knowledge of the immune/inflammatory pathways which characterize the pathophysiology of RA provided the link between inflammation and bone loss via a complex network of bone cells; T and B cells; proinflammatory cytokines such as TNF-α, IL-1, IL-6, IL-17, and IL 23; co-stimulator molecules; signaling pathways, including both the RANK ligands – RANK and osteoprogeterin (RANKL/RANK/OPG); and Wnt inhibitor dickkopf-1 Wnt (DKK-1) signaling.Citation58Citation60

As a consequence, it has been postulated that the novel targeted biologic therapies, which are characterized by powerful anti-inflammatory activity, would reduce both systemic bone loss and fracture incidence.Citation58,Citation61,Citation62

Several cross-sectional and prospective studies evaluated the effect of biologics on bone mineral density (BMD) and/or bone turnover markers (BTMs) in RA with conflicting results. Most of these studies have been conducted with anti-TNFs, whereas there are very few studies with non-anti-TNF-targeted TCZ, RTX, and ABA.

Anti-TNFs and bone mass

TNF-α plays a central role in the physiopathology of bone loss by decreasing osteoblast activity and survival and increasing osteoclast differentiation, recruitment, and activity.Citation58Citation60 Consequently, these findings suggest a beneficial effect of anti-TNFs also in preventing systemic bone loss.

Krieckaert et alCitation63 documented that ADA treatment over 4 years arrested bone loss in the lumbar spine in 184 patients with severe RA, whereas hip BMD continued to decrease. A study evaluating the effect of synthetic DMARDs and biologics on systemic bone loss in RA patients who were followed for 10 years showed that a more aggressive strategy, including anti-TNFs, was effective in reducing the rate of bone loss.Citation64 By contrast, a review by Kawai et alCitation65 concluded that anti-TNFs were not effective in preventing bone loss in RA. Interestingly, data deriving from a recent meta-analysis on the effects of anti-TNFs on BMD in RA and AS trialsCitation66 showed a significant improvement of both lumbar spine and hip BMD only in AS patients. Recent data deriving from clinical studies between 2005 and 2014 added evidence that TNF-α inhibitors are able to arrest systemic bone loss as assessed by BMD and BTMs in RA.Citation67 The beneficial effects of anti-TNFs against bone loss have been confirmed in other reviews.Citation62,Citation68

IL-6 blockade

The IL-6 pathway is one of the major contributing factors to the disruption of bone metabolism in immuno-mediated inflammatory diseases.Citation59Citation61 IL-6 is a pleiotropic cytokine strongly connected to osteoclast physiology, as it interacts synergistically with IL-1 and TNF-α to promote the differentiation of osteoclast precursors (OCPs), to prolong the survival and increase the activity of mature osteoclasts. Moreover, this cytokine plays a critical role in the generation of proinflammatory TH17 cells, thereby enhancing IL-17 levels.Citation60

The effect of TCZ on BTMs has been investigated in some prospective studies of RA patients. A rapid and significant increase of bone-formation markers (BFMs) associated with a significant decrease of bone-resorption markers (BRMs) followed the initiation of TCZ combination therapy with MTX in 416 patients with active RA.Citation69 In addition, in the RADIATE study, TCZ–MTX treatment significantly decreased BRMs and increased BFMs, although not significantly, in RA patients.Citation70 A critical role of TCZ on bone metabolism was also suggested by a pilot study which, in a smaller sample of RA patients (22 women), documented that the IL-6 blockade increased the OPG/RANKL ratio and decreased DKK-1 levels after 2 months of treatment.Citation71 More recently, the effect of TCZ has been evaluated on BMD in patients with MTX-resistant active RA.Citation72 Interestingly, TCZ stably maintained the BMD in patients with normal baseline BMD and increased BMD in osteopenic patients at baseline. In contrast, a 1-year open prospective study in patients with active RA receiving TCZ and MTX did not show significant changes in BMD.Citation73 However, a significant decrease of serum levels of DKK-1 and a significant increase of BFMs were associated with TCZ–MTX treatment.

Inhibition of B and T cells

Together with activated T cells, B cells highly express RANKL and IL-6, thereby contributing to osteoclastogenesis.Citation59 Moreover, B cells differentiate into plasma cells, which in turn inhibit bone formation through the expression of the DKK-1.Citation59

Clinical studies evaluating the effect of RTX on systemic bone loss are not available. Nevertheless, as a consequence of the critical role played by B cells on systemic bone homeostasis, it can be speculated that RTX can positively influence bone mass.Citation54 Boumans et alCitation74 showed that 12-month treatment with RTX was associated with an increased OPG/RANKL ratio in the serum of 28 RA patients. In contrast, in a prospective study with a follow-up of 15 months that evaluated 13 RA patients, a non-significant decrease of RANKL was observed after RTX treatment.Citation75 Wheater et alCitation76 documented a significant decrease of BRMs associated with a significant increase of BFMs after 6 months of treatment with RTX in 46 RA patients.

So far, there are no clinical studies with BMD and/or BTs as primary outcomes in RA patients treated with ABA. CTLA-4 is a T-cell-associated antigen which competes with CD28 to bind to CD80/CD86, which is a potent costimulatory signal for T cells and OCPs.Citation58,Citation61 In a model of TNF-α-induced arthritis, CTLA-4 dose-dependently and directly inhibited osteoclast differentiation and maturation by binding to their precursor cells.Citation77 Recently, Bozec et alCitation78 reported that the binding of CTLA-4 to CD80/86 in OCPs induced their apoptosis. Thus, these data may suggest that ABA could exert a beneficial effect on systemic bone loss.Citation60,Citation67

Biologics and fractures

To date, the effect of biologics on fracture risk remains uncertain. Data from the CORRONA registry, which evaluated 11,429 RA patients treated with synthetic DMARDs or TNF-α inhibitors as monotherapy or synthetic DMARDs in combination with TNF-α inhibitors, showed a decrease in overall fracture risk associated with anti-TNF treatment when compared with other treatments.Citation79 In contrast, Kim et alCitation80 found a similar risk of non-vertebral fractures in RA patients treated with anti-TNFs as monotherapy or in combination with a synthetic DMARD, MTX alone, or another synthetic DMARD alone. Similar findings resulted from a nested case– control study of 1,515 RA patients treated with biologics, mostly anti-TNFs, and 6,023 controls.Citation81 Furthermore, the risk of combined non-vertebral fractures did not differ between initiators of a synthetic agent and an anti-TNF in a large retrospective cohort of patients with RA and spondyloarthritis.Citation82 Recently, three reviews which evaluated the anti-fracture activity of biologics, chiefly anti-TNFs, failed to demonstrate a clinically meaningful effect.Citation65,Citation67,Citation68

Biologics and PD

The relationship between autoimmune rheumatic disorders and PD has been chiefly investigated in patients with RA with a focus on the epidemiology, pathogenic interplay, effect of coexisting PD on the clinical course of RA and response to biologic therapies, and the effects of biologics on PD.

Confirming previous reports,Citation83Citation89 in a case–control study of 287 patients with established RA, there was a higher frequency of PD as compared with 330 matched controls (35%; unadjusted OR 1.49; 95% CI 1.06, 2.11; P=0.02). The difference was more pronounced in anti-citrullinated protein antibodies (ACPA)-positive cases (37%; unadjusted OR 1.65; 95% CI 1.15, 2.36; P=0.006). The association persisted significantly and more frequently after multivariate analysis adjustment in ACPA-positive patients (OR 1.59; 95% CI 1.01, 2.49; P=0.043).Citation90 Two large population-based studies confirmed these findings,Citation91,Citation92 whereas another large-scale epidemiologic research study did not.Citation93

These conflicting results were observed in patients with longstanding RA, where two studies of new-onset RA confirmed the association with PD.Citation88,Citation94 The relationship between RA and PD seems to be strengthened by the circumstance that the two inflammatory conditions share increased TNF production.Citation95

Although controversial, these epidemiologic results suggest common pathogenic pathways between RA and PD; moreover, the role of the oral microbiome as a triggering factor for RA has been especially emphasized.Citation96,Citation97 In particular, a component of the oral microbiome – Porphyromonas gingivalis – seems to play an important pathogenic role in PD, ACPA production, and RA development.Citation98,Citation99

Mutual effects of biologic therapy and PD

The possible connection between the two inflammatory conditions seems to be confirmed by the negative influx on biologic therapy effectiveness exerted by coexistent PD in patients with RA. In a limited series of 18 RA patients with PD, a significant lower efficacy of anti-TNF therapy was observed in RA patients with PD;Citation100 furthermore, a significant association between the coexistence of PD and the risk of ETN discontinuation resulted in a large population-based cohort of 3,359 patients (HR 1.27; 95% CI 1.01, 1.60).Citation101 Conversely, several studies reported the beneficial effect of non-surgical PD treatment on RA severity.Citation102Citation105

The effects of biologics on PD have been chiefly investigated in patients with RA receiving anti-TNFs. Overall, the majority of the studies evidenced a reduction of gingival inflammation with improvement of parameters of PD severity.Citation106Citation110 Furthermore, similar results were observed in patients with ankylosing spondylitis.Citation111 However, the strength of this evidence is limited by the open-label design of the studies and by the limited number of patients evaluated.

Fertility and sexuality in RA patients treated with biologics

Dealing with these topics implies a consideration of several aspects, including the role exerted by RA itself, the effects of biologic-combined therapies, and the direct effects of biologics. Although often less regarded, the fallout on patients’ psychological status may be detrimental, especially if the RA-induced depressive condition in at least one third of the patients is taken in account.Citation112

With regard to fertility in RA as reported in other reports,Citation113 a recent study observed unexplained subfertility and anovulation in 48% and 28%, respectively, of 178 women.Citation114 Moreover, RA itself impairs the sexual function in up to 70% of women,Citation115 with a direct correlation with the disease activity.Citation116 Both fertility status and sexual function in males have been poorly investigated, with inconclusive results. However, reduced levels of testosterone have been detected in a low number of cohorts of males with RA.Citation117,Citation118 Even data on sexual function in males with RA are extremely limited, with erectile dysfunction being reported in ten (33%) out of 31 males in only one study.Citation117 Among the traditional immuno-suppressants employed for the treatment of RA, sulfasalazine has been proven to impair fertility,Citation118 whereas data on MTX are unavailable.Citation119,Citation120 No impact on fertility has been reported for cyclosporine and azathioprine.Citation119

Anti-TNFs have been extensively investigated for use in pregnancy; however, to the best of our knowledge, studies to assess their effects on fertility and sexual function in patients with RA are lacking. Data from patients with ankylosing spondylitis indicate that anti-TNFs do not impair sperm quality and fertilityCitation121 and may improve sexual dysfunction in males.Citation122 To date, no data on fertility and sexual dysfunction are available for non-anti-TNF-targeted biologics.

Discussion

Available recommendations for the management of patients with RA provide important indications for the appropriate use of biologics, their efficacy and safety, and correct patient follow-up.Citation123,Citation124 Similarly, systematic reviews and meta-analyses have been chiefly focused on the same topics.Citation125 However, the fallout on real-life clinical practice of the issues discussed in the present paper and the consequent management of patients have been rarely evaluated. In most cases, rheumatologists carefully address and measure the clinical response, but often neglect other patient characteristics that, in turn, may reduce or complicate the effects of therapeutic interventions.

As shown in , the ITABIO task force has formulated several evidence-based statements useful for improving the outcome of patients with RA.

Table 2 Frequently disregarded issues in the management of patients with rheumatoid arthritis: evidence-based ITABIO statements useful for the rheumatology clinical practice

Focusing on immunogenicity, a consistent number of studies have evaluated the immune mechanisms and the effects on efficacy of anti-TNFs; however, in most cases, no suggestions for clinical practice were formulated.Citation122 In this regard, when anti-TNFs are indicated, Statement 1 suggests combination therapy with MTX; otherwise, non-anti-TNFs are preferable. In the case of anti-TNF secondary failure, switching to a non-anti-TNF is advisable.Citation127,Citation128

Statement 2 provides indications for the suspect and diagnosis of infections in patients receiving biologics, with the suggestion to treat patients with high infection risk with a non-anti-TNF biologic.

As indicated in statement 3, although LLS is infrequent, rheumatologists should be aware of the possible occurrence of this syndrome in a patient taking anti-TNFs who develops fever and skin and articular complaints associated with a positive ANA titer. After anti-TNF interruption, at LLS recovery, a non-anti-TNF-targeted biologic is advisable.

Statement 4 reflects the existing evidence, and the key message for clinicians is to drive the biologic choice toward an anti-TNF in the case of a female patient with adjunctive risk factors for osteoporosis.

As pointed out in Statement 5, the connection between PD and RA requires further investigation; nevertheless, rheumatologists should consider the periodontal status of the patient and motivate him/her to undergo appropriate dental care.

Finally, Statement 6 explores a delicate issue that, often, is omitted by the patient due to reserve or shame in discussing it. However, to provide complete assistance, rheumatologists should ask the patient about his/her limitations and counsel psychiatric support.

Conclusion

In a correct and complete evaluation of the patient with RA, the discussed issues are often disregarded in clinical practice; however, an “in toto” evaluation of all patient features may drive the choice of biologic therapy and improve the outcome. We believe that information summarized in this article would be useful to clinicians for a more comprehensive patient management.

Disclosure

The authors report no conflicts of interest in this work.

References

  • CantiniFNiccoliLNanniniCItalian board for the TAilored BIOlogic therapy (ITABIO). Tailored first-line biologic therapy in patients with rheumatoid arthritis, spondyloarthritis, and psoriatic arthritisSemin Arthritis Rheum20164551953226607440
  • MokCCTsaiWCChenDYWeiJCCImmunogenicity of anti-TNF biologic agents in the treatment of rheumatoid arthritisExpert Opin Biol Ther201616220121126560845
  • MeroniPLValentiniGAyalaFCattaneoAValesiniGNew strategies to address the pharmacodynamics and pharmacokinetics of tumor necrosis factor (TNF) inhibitors: A systematic analysisAutoimmun Rev201514981282925985765
  • GunnGRSealeyDCFJamaliFMeibohmBGhoshSShankarGFrom the bench to clinical practice: understanding the challenges and uncertainties in immunogenicity testing for biopharmaceuticalsClin Exp Immunol2016184213714626597698
  • SethuSGovindappaKAlhaidariMPirmohamedMParkKSathishJImmunogenicity to biologics: mechanisms, prediction and reductionArch Immunol Ther Exp2012605331344
  • JullienDPrinzJCNestleFOImmunogenicity of biotherapy used in psoriasis: the science behind the scenesJ Invest Dermatol20151351313825120005
  • RatanjiKDDerrickJPDearmanRJKimberIImmunogenicity of therapeutic proteins: Influence of aggregationJ Immunotoxicol20141129910923919460
  • GoinsCLChappellCPShashidharamurthyRSelvarajPJacobJImmune complex-mediated enhancement of secondary antibody responsesJ Immunol2010184116293629820439912
  • BatistaFDHarwoodNEThe who, how and where of antigen presentation to B cellsNat Rev Immunol200991152719079135
  • WadhwaMThorpeRStrategies and Assays for the Assessment of Unwanted ImmunogenicityJ Immunotoxicol20063311512118958691
  • StrandVBalsaAAl-SalehJImmunogenicity of Biologics in Chronic Inflammatory Diseases: A Systematic ReviewBioDrugs201731429931628612180
  • KrieckaertCRispensTWolbinkGImmunogenicity of biological therapeutics: from assay to patientCurr Opin Rheumatol20122430631122388644
  • SchaeverbekeTTruchetetM-EKostineMBarnetcheTBannwarthBRichezCImmunogenicity of biologic agents in rheumatoid arthritis patients: lessons for clinical practiceRheumatology201655221022026268816
  • GarcêsSDemengeotJBenito-GarciaEThe immunogenicity of anti-TNF therapy in immune-mediated inflammatory diseases: a systematic review of the literature with a meta-analysisAnn Rheum Dis201372121947195523223420
  • ManeiroJRSalgadoEGomez-ReinoJJImmunogenicity of Monoclonal Antibodies Against Tumor Necrosis Factor Used in Chronic Immune-Mediated Inflammatory ConditionsJAMA Intern Med2013173151416142823797343
  • ThomasSSBorazanNBarrosoNComparative Immunogenicity of TNF Inhibitors: Impact on Clinical Efficacy and Tolerability in the Management of Autoimmune Diseases. A Systematic Review and Meta-AnalysisBioDrugs201529424125826280210
  • MootsRJXavierRMMokCCThe impact of anti-drug antibodies on drug concentrations and clinical outcomes in rheumatoid arthritis patients treated with adalimumab, etanercept, or infliximab: Results from a multinational, real-world clinical practice, non-interventional studyPLoS One2017124e017520728448562
  • BurmesterGRChoyEKivitzALow immunogenicity of tocilizumab in patients with rheumatoid arthritisAnn Rheum Dis20177661078108528007755
  • BenucciMMeacciFGrossiVCorrelations between immunogenicity, drug levels, and disease activity in an Italian cohort of rheumatoid arthritis patients treated with tocilizumabBiologics201610535827041992
  • SigauxJHamzeMDaienCImmunogenicity of tocilizumab in patients with rheumatoid arthritisJoint Bone Spine2017841394527369643
  • ListingJGerholdKZinkAThe risk of infections associated with rheumatoid arthritis, with its comorbidity and treatmentRheumatology2013521536123192911
  • MinozziSBonovasSLytrasTRisk of infections using anti-TNF agents in rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis: a systematic review and meta-analysisExpert Opin Drug Saf201615sup1113427924643
  • RoachDRBeanAGDDemangelCFranceMPBriscoeHBrittonWJTNF regulates chemokine induction essential for cell recruitment, granuloma formation, and clearance of mycobacterial infectionJ Immunol200216894620462711971010
  • KooSMartyFMBadenLRInfectious Complications Associated with Immunomodulating Biologic AgentsInfect Dis Clin North Am201024228530620466271
  • CantiniFNiccoliLGolettiDTuberculosis Risk in Patients Treated with Non-Anti-Tumor Necrosis Factor- (TNF-) Targeted Biologics and Recently Licensed TNF-Inhibitors: Data from Clinical Trials and National RegistriesJ Rheumatol Suppl201491566424789001
  • WallisRSBroderMWongJLeeAHoqLReactivation of latent granulomatous infections by infliximabClin Infect Dis200541Supplement_3S194S19815983899
  • TorunerMLoftusEVHarmsenWSRisk factors for opportunistic infections in patients with inflammatory bowel diseaseGastroenterology2008134492993618294633
  • CantiniFBocciaSGolettiDHBV Reactivation in Patients Treated with Antitumor Necrosis Factor-Alpha (TNF-α) Agents for Rheumatic and Dermatologic Conditions: A Systematic Review and Meta-AnalysisInt J Rheumatol20142014292683625114684
  • StrangfeldAListingJHerzerPRisk of Herpes Zoster in Patients With Rheumatoid Arthritis Treated With Anti–TNF-α AgentsJAMA2009301773774419224750
  • AliTKaithaSMahmoodSFtesiAStoneJBronzeMSClinical use of anti-TNF therapy and increased risk of infectionsDrug Health Patient Saf201357999
  • AsklingJForedCMBrandtLTime-dependent increase in risk of hospitalisation with infection among Swedish RA patients treated with TNF antagonistsAnn Rheum Dis200766101339134417261532
  • WeinblattMESchiffMValenteRHead-to-head comparison of subcutaneous abatacept versus adalimumab for rheumatoid arthritis: Findings of a phase IIIb, multinational, prospective, randomized studyArthritis Rheum2013651283823169319
  • ZinkAStrangfeldASchneiderMEffectiveness of tumor necrosis factor inhibitors in rheumatoid arthritis in an observational cohort study: Comparison of patients according to their eligibility for major randomized clinical trialsArthritis Rheum200654113399340717075823
  • AaltonenKJYlikyläSTuulikki JoensuuJEfficacy and effectiveness of tumour necrosis factor inhibitors in the treatment of rheumatoid arthritis in randomized controlled trials and routine clinical practiceRheumatology201756572573528064209
  • SinghJAInfections With Biologics in Rheumatoid Arthritis and Related Conditions: a Scoping Review of Serious or Hospitalized Infections in Observational StudiesCurr Rheumatol Rep201618106127613285
  • RhodesAEvansLEAlhazzaniWSurviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock: 2016Intensive Care Med201743330437728101605
  • SingerMDeutschmanCSSeymourCWThe Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3)JAMA2016315880181026903338
  • SeymourCWLiuVXIwashynaTJAssessment of Clinical Criteria for Sepsis: For the Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3)JAMA2016315876277426903335
  • LauplandKBIncidence of bloodstream infection: a review of population-based studiesClin Microbiol Infect201319649250023398633
  • BuettiNAtkinsonAMarschallJKronenbergASwiss Centre for Antibiotic Resistance (ANRESIS). Incidence of bloodstream infections: a nationwide surveillance of acute care hospitals in Switzerland 2008–2014BMJ Open20177e013665
  • ChandraRKNutrition, immunity and infection: From basic knowledge of dietary manipulation of immune responses to practical application of ameliorating suffering and improving survivalProc Natl Acad Sci U S A1996932514304143078962043
  • Fernandez-MartinezSCortesXBorrás-BlascoJGracia-PérezACasteráMDEEffectiveness of a systematic vaccination program in patients with autoimmune inflammatory disease treated with anti-TNF alpha drugsExpert Opin Biol Ther201616111317132227537179
  • WilliamsELGadolaSEdwardsCJAnti-TNF-induced lupusRheumatology200948771672019416947
  • Pereira VazJLFernandesVNogueiraFArnobioAMLevyRAInfliximab induced autoantibodies. A multicenter studyClin Rheumatol20163532533226676808
  • ChimentiMSSpinelliFRGiuntaAEmergence of antinuclear antibodies in psoriatic patients treated with Infliximab: personal experience and literature reviewDrug Dev Res201475Suppl 1S61S6325381980
  • Ramos-CasalsMBrito-ZerónPSotoM-JCuadradoM-JKhamashtaMAAutoimmune diseases induced by TNF-targeted therapiesBest Pract Res Clin Rheumatol200822584786119028367
  • KatzUZandman-GoddardGDrug-induced lupus: An updateAutoimmun Rev2010101465020656071
  • De BandtMSibiliaJLe LoëtXSystemic lupus erythematosus induced by anti-tumour necrosis factor alpha therapy: a French national surveyArthritis Res Ther200573R545R55115899041
  • WetterDADavisMDPLupus-Like Syndrome Attributable to Anti – Tumor Necrosis Factor α Therapy in 14 Patients During an 8-Year Period at Mayo ClinicMayo Clin Proc2009841197998419880688
  • CostaMFSaidNRZimmermannBDrug-Induced Lupus due to Anti-Tumor Necrosis Factor α AgentsSemin Arthritis Rheum200837638138717977585
  • VannucchiVGrazziniMPieralliFGiannottaMBiagioniCNozzoliCAdalimumab-induced lupus erythematosus with central nervous system involvement in a patient with Crohn’s diseaseJ Gastrointestin Liver Dis201120220120321725519
  • AringerMSmolenJSTherapeutic blockade of TNF in patients with SLE – Promising or crazy?Autoimmun Rev201211532132521619949
  • MoscaMTaniCFiliceMETNF-alpha inhibitors in Systemic Lupus Erythematosus. A case report and a systematic literature reviewMod Rheumatol201525464264524252029
  • EmerJJFrankelAZeichnerJAA practical approach to monitoring patients on biological agents for the treatment of psoriasisJ Clin Aesthet Dermatol2010382026
  • DingTLedinghamJLuqmaniRBSR and BHPR rheumatoid arthritis guidelines on safety of anti-TNF therapiesRheumatology201049112217221920837498
  • SinigagliaLNervettiAMelaQA multicenter cross sectional study on bone mineral density in rheumatoid arthritis. Italian Study Group on Bone Mass in Rheumatoid ArthritisJ Rheumatol200027112582258911093437
  • OkanoTInuiKTadaMHigh frequency of vertebral fracture and low bone quality in patients with rheumatoid arthritis – Results from TOMORROW studyMod Rheumatol201727339840427484855
  • SchettGStachCZwerinaJVollRMangerBHow antirheumatic drugs protect joints from damage in rheumatoid arthritisArthritis Rheum200858102936294818821703
  • ShawATGravalleseEMMediators of inflammation and bone remodeling in rheumatic diseaseSemin Cell Dev Biol20164921026481971
  • AmarasekaraDSYuJRhoJBone Loss Triggered by the Cytokine Network in Inflammatory Autoimmune DiseasesJ Immunol Res2015 2015201583212726065006
  • RedlichKSmolenJSInflammatory bone loss: pathogenesis and therapeutic interventionNat Rev Drug Discov201211323425022378270
  • DimitroulasTNikasSNTrontzasPKitasGDBiologic therapies and systemic bone loss in rheumatoid arthritisAutoimmun Rev2013121095896623542506
  • KrieckaertCLMNurmohamedMTWolbinkGLemsWFChanges in bone mineral density during long-term treatment with adalimumab in patients with rheumatoid arthritis: a cohort studyRheumatology201352354755323221326
  • HaugebergGHelgetveitKBFørreØGarenTSommersethHPrøvenAGeneralized bone loss in early rheumatoid arthritis patients followed for ten years in the biologic treatment eraBMC Musculoskelet Disord201415128925182527
  • KawaiVKSteinCMPerrienDSGriffinMREffects of anti-tumor necrosis factor α agents on boneCurr Opin Rheumatol201224557658522810364
  • SiuSHaraouiBBissonnetteRMeta-analysis of tumor necrosis factor inhibitors and glucocorticoids on bone density in rheumatoid arthritis and ankylosing spondylitis trialsArthritis Care Res2015676754764
  • ManaraMSinigagliaLBone and TNF in rheumatoid arthritis: clinical implicationsRMD Open20151Suppl 1e00006526557382
  • ZerbiniCAFClarkPMendez-SanchezLBiologic therapies and bone loss in rheumatoid arthritisOsteoporos Int201728242944627796445
  • GarneroPThompsonEWoodworthTSmolenJSRapid 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 aloneArthritis Rheum2010621334320039425
  • KarsdalMASchettGEmeryPIL-6 receptor inhibition positively modulates bone balance in rheumatoid arthritis patients with an inadequate response to anti-tumor necrosis factor therapy: biochemical marker analysis of bone metabolism in the tocilizumab RADIATE study (NCT00106522)Semin Arthritis Rheum201242213113922397953
  • TerposEFragiadakiKKonstaMBratengeierCPapatheodorouASfikakisPPEarly effects of IL-6 receptor inhibition on bone homeostasis: a pilot study in women with rheumatoid arthritisClin Exp Rheumatol201129692192522032557
  • KumeKAmanoKYamadaSThe effect of tocilizumab on bone mineral density in patients with methotrexate-resistant active rheumatoid arthritisRheumatology201453590090324441151
  • BriotKRouanetSSchaeverbekeTThe effect of tocilizumab on bone mineral density, serum levels of Dickkopf-1 and bone remodeling markers in patients with rheumatoid arthritisJoint Bone Spine201582210911525557658
  • BoumansMJHThurlingsRMYeoLRituximab abrogates joint destruction in rheumatoid arthritis by inhibiting osteoclastogenesisAnn Rheum Dis201271110811322072013
  • HeinGEidnerTOelznerPInfluence of Rituximab on markers of bone remodeling in patients with rheumatoid arthritis: a prospective open-label pilot studyRheumatol Int201131226927220661741
  • WheaterGHoganVETengYKOSuppression of bone turnover by B-cell depletion in patients with rheumatoid arthritisOsteoporosis International201122123067307221625887
  • AxmannRHermanSZaissMCTLA-4 directly inhibits osteoclast formationAnn Rheum Dis200867111603160918203760
  • BozecAZaissMMKagwiriaRT cell costimulation molecules CD80/86 inhibit osteoclast differentiation by inducing the IDO/tryptophan pathwaySci Transl Med20146235235ra60
  • CoulsonKAReedGGilliamBEKremerJMPepmuellerPHFactors influencing fracture risk, T score, and management of osteoporosis in patients with rheumatoid arthritis in the Consortium of Rheumatology Researchers of North America (CORRONA) registryJ Clin Rheumatol200915415516019363452
  • KimSYSchneeweissSLiuJSolomonDHEffects of disease-modifying antirheumatic drugs on nonvertebral fracture risk in rheumatoid arthritis: A population-based cohort studyJ Bone Miner Res201227478979622162140
  • RoussyJ-PBessetteLBernatskySRahmeELachaineJBiologic disease-modifying anti-rheumatic drugs and the risk of non-vertebral osteoporotic fractures in patients with rheumatoid arthritis aged 50 years and overOsteoporos Int20132492483249223504029
  • KawaiVKGrijalvaCGArbogastPGInitiation of tumor necrosis factor α antagonists and risk of fractures in patients with selected rheumatic and autoimmune diseasesArthritis Care Res201365710851094
  • KässerURGleissnerCDehneFMichelAWillershausen-ZönnchenBBoltenWWRisk for periodontal disease in patients with longstanding rheumatoid arthritisArthritis Rheum19974012224822519416864
  • MercadoFBMarshallRIKlestovACBartoldPMRelationship Between Rheumatoid Arthritis and PeriodontitisJ Periodontol200172677978711453241
  • PischonNPischonTKrögerJAssociation among rheumatoid arthritis, oral hygiene, and periodontitisJ Periodontol200879697998618533773
  • GaribBTQaradaxiSSTemporomandibular joint problems and periodontal condition in rheumatoid arthritis patients in relation to their rheumatologic statusJ Oral Maxillofac Surg201169122971297821764200
  • SmitMWestraJVissinkADoornbosvan der MeerBBrouwerEvan WinkelhoffAPeriodontitis in established rheumatoid arthritis patients: a cross-sectional clinical, microbiological and serological studyArthritis Res Ther2012145R22223075462
  • ScherJUUbedaCEquindaMPeriodontal disease and the oral microbiota in new-onset rheumatoid arthritisArthritis Rheum201264103083309422576262
  • PotikuriDDannanaKCKanchinadamSPeriodontal disease is significantly higher in non-smoking treatment-naive rheumatoid arthritis patients: results from a case-control studyAnn Rheum Dis20127191541154422875903
  • JosephRRajappanSNathSGPaulBJAssociation between chronic periodontitis and rheumatoid arthritis: a hospital-based case–control studyRheumatol Int201333110310922228465
  • MikulsTRPayneJBYuFPeriodontitis and Porphyromonas gingivalis in Patients With Rheumatoid ArthritisArthritis Rheumatol20146651090110024782175
  • ArkemaEVKarlsonEWCostenbaderKHA prospective study of periodontal disease and risk of rheumatoid arthritisJ Rheumatol20103791800180420595268
  • DemmerRTMolitorJAJacobsDRMichalowiczBSPeriodontal disease, tooth loss and incident rheumatoid arthritis: results from the First National Health and Nutrition Examination Survey and its epidemio-logical follow-up studyJ Clin Periodontol20113811998100622092471
  • ErikssonKNiseLKatsAPrevalence of Periodontitis in Patients with Established Rheumatoid Arthritis: A Swedish Population Based Case-Control StudyPLoS One2016115e015595627203435
  • WolffBBergerTFreseCOral status in patients with early rheumatoid arthritis: a prospective, case-control studyRheumatology201453352653124273047
  • JavedFAhmedHBMikamiTAlmasKRomanosGEAl-HezaimiKCytokine profile in the gingival crevicular fluid of rheumatoid arthritis patients with chronic periodontitisJ Investig Clin Dent20145118
  • ScherJUBretzWAAbramsonSBPeriodontal disease and subgingival microbiota as contributors for rheumatoid arthritis pathogenesisCurr Opin Rheumatol201426442442924807405
  • BruscaSBAbramsonSBScherJUMicrobiome and mucosal inflammation as extra-articular triggers for rheumatoid arthritis and autoimmunityCurr Opin Rheumatol201426110110724247114
  • AlgoodHMLinPLYankuraDJonesAChanJFlynnJLTNF influences chemokine expression of macrophages in vitro and that of CD11b+ cells in vivo during Mycobacterium tuberculosis infectionJ Immunol2004172116846685715153503
  • AraújoVMAMeloIMLimaVRelationship between Periodontitis and Rheumatoid Arthritis: Review of the LiteratureMediators Inflamm2015201525907426347200
  • SavioliCRibeiroACFabriGMPersistent periodontal disease hampers anti-tumor necrosis factor treatment response in rheumatoid arthritisJ Clin Rheumatol201218418018422647860
  • ChenHHChenDYLaiKLPeriodontitis and etanercept discontinuation risk in anti-tumor necrosis factor-naive rheumatoid arthritis patients: a nationwide population-based cohort studyJ Clin Rheumatol201319843243824263145
  • OrtizPBissadaNFPalomoLPeriodontal therapy reduces the severity of active rheumatoid arthritis in patients treated with or without tumor necrosis factor inhibitorsJ Periodontol200980453554019335072
  • BıyıkoğluBBuduneliNAksuKPeriodontal therapy in chronic periodontitis lowers gingival crevicular fluid interleukin-1beta and DAS28 in rheumatoid arthritis patientsRheumatol Int201333102607261623728274
  • ErciyasKSezerUUstünKEffects of periodontal therapy on disease activity and systemic inflammation in rheumatoid arthritis patientsOral Dis201319439440022998534
  • OkadaMKobayashiTItoSPeriodontal treatment decreases levels of antibodies to Porphyromonas gingivalis and citrulline in patients with rheumatoid arthritis and periodontitisJ Periodontol20138412e74e8423701010
  • MayerYElimelechRBalbir-GurmanABraun-MoscoviciYMachteiEEPeriodontal condition of patients with autoimmune diseases and the effect of anti-tumor necrosis factor-α therapyJ Periodontol201384213614222524332
  • KadkhodaZAmirzargarAEsmailiZVojdanianMAkbariSEffect of TNF-α Blockade in Gingival Crevicular Fluid on Periodontal Condition of Patients with Rheumatoid ArthritisIran J Immunol201613319720327671511
  • KobayashiTYokoyamaTItoSPeriodontal and serum protein profiles in patients with rheumatoid arthritis treated with tumor necrosis factor inhibitor adalimumabJ Periodontol201485111480148824857321
  • HanJYReynoldsMAEffect of anti-rheumatic agents on periodontal parameters and biomarkers of inflammation: a systematic review and meta-analysisJ Periodontal Implant Sci201242131222413068
  • FabriGMPereiraRMSavioliCPeriodontitis Response to Anti-TNF Therapy in Ankylosing SpondylitisJ Clin Rheumatol201521734134526398459
  • MatchamFRaynerLSteerSHotopfMThe prevalence of depression in rheumatoid arthritis: a systematic review and meta-analysisRheumatology201352122136214824003249
  • OstensenMRheumatoid arthritis: The effect of RA and medication on female fertilityNat Rev Rheumatol201410951851925003768
  • BrouwerJFleurbaaijRHazesJMWDolhainRLavenJSESub-fertility in Women With Rheumatoid Arthritis and the Outcome of Fertility AssessmentsArthritis Care Res201769811421149
  • CostaTFSilvaCRMunizLFMotaLMPrevalence of sexual dysfunction among female patients followed in a Brasília Cohort of early rheumatoid arthritisRev Bras Reumatol201555212313225559062
  • BrouwerJHazesJMLavenJSDolhainRJFertility in women with rheumatoid arthritis: influence of disease activity and medicationAnn Rheum Dis201574101836184124833784
  • GordonDBeastallGHThomsonJASturrockRDAndrogenic status and sexual function in males with rheumatoid arthritis and ankylosing spondylitisQ J Med1986602316716793094090
  • CutoloMBalleariEGiustiMMonachesiMAccardoSSex hormone status of male patients with rheumatoid arthritis: evidence of low serum concentrations of testosterone at baseline and after human chorionic gonadotropin stimulation. Sex hormone status of male patients with rheumatoid arthritis: evidence of low serum concentrations of testosterone at baseline and after human chorionic gonadotropin stimulationArthritis Rheum198831131413173140823
  • LeroyCRigotJMLeroyMImmunosuppressive drugs and fertilityOrphanet J Rare Dis20151013626490561
  • GutierrezJCHwangKThe toxicity of methotrexate in male fertility and paternal teratogenicityExpert Opin Drug Metab Toxicol2017131515827590039
  • RamondaRForestaCOrtolanAInfluence of tumor necrosis factor α inhibitors on testicular function and semen in spondyloarthritis patientsFertil Steril2014101235936524332378
  • OhJSHeoHMKimYGLeeSGLeeCKYooBThe effect of anti-tumor necrosis factor agents on sexual dysfunction in male patients with ankylosing spondylitis: a pilot studyInt J Impot Res200921637237519759542
  • SmolenJSLandewéRBijlsmaJEULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 updateAnn Rheum Dis201776696097728264816
  • SinghJASaagKGBridgesSL2015 American College of Rheumatology Guideline for the Treatment of Rheumatoid ArthritisArthritis Rheumatol2016681126
  • TarpSEric FurstDBoersMRisk of serious adverse effects of biological and targeted drugs in patients with rheumatoid arthritis: a systematic review meta-analysisRheumatology201756341742528013201
  • AtkinsDBestDBrissPAGrading quality of evidence and strength of recommendationsBMJ20043287454149015205295
  • CantiniFNiccoliLNanniniCSecond-line biologic therapy optimization in rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitisSemin Arthritis Rheum201747218319228413099
  • FavalliEGRaimondoMGBeccioliniACrottiCBiggioggeroMCaporaliRThe management of first-line biologic therapy failures in rheumatoid arthritis: Current practice and future perspectivesAutoimmun Rev201716121185119529037899