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

Tumor necrosis factor-alpha inhibitors: Induction of antibodies, autoantibodies, and autoimmune diseases

Tumor necrosis factor-alpha inhibitors: Induction of antibodies, autoantibodies, and autoimmune diseases
Literature review current through: Jan 2024.
This topic last updated: Jul 30, 2021.

INTRODUCTION — Targeted inhibitors of tumor necrosis factor (TNF)-alpha are widely used in the treatment of a number of inflammatory conditions, including rheumatoid arthritis; axial and peripheral spondyloarthritis; juvenile idiopathic arthritis; inflammatory bowel diseases; plaque psoriasis, pustular psoriasis, and psoriatic arthritis; sarcoidosis; noninfectious uveitis; hidradenitis suppurativa; and others. However, prolonged use of these agents has been associated in some patients with the development of anti-drug antibodies (ADA), which may promote adverse effects and diminish drug efficacy and cost-effectiveness. Induction of autoimmunity, including both autoantibodies and, much less often, clinically evident autoimmune disease, may also occur during treatment.

Adverse effects associated with the use of biologic agents that inhibit TNF include injection site and acute and delayed infusion reactions, cytopenias, infections, autoimmune and demyelinating diseases, heart failure, malignancy, and pulmonary, liver, and skin diseases. These potentially serious risks are among the factors included in the risk/benefit assessment made when deciding whether to use one of these agents in an individual patient.

TNF inhibitor-related ADA formation, autoantibody formation, and the development of autoimmune diseases are discussed here. Other major complications of anti-TNF therapy, such as injection site and infusion reactions, tuberculosis and other infections, malignancy, and other adverse effects, are described separately. (See "Tumor necrosis factor-alpha inhibitors: An overview of adverse effects" and "Risk of mycobacterial infection associated with biologic agents and JAK inhibitors" and "Tumor necrosis factor-alpha inhibitors: Bacterial, viral, and fungal infections" and "Tumor necrosis factor-alpha inhibitors: Risk of malignancy".)

ANTI-DRUG ANTIBODIES — The formation of anti-drug antibodies (ADA), especially drug-neutralizing ADA, is a concern with biologic tumor necrosis factor (TNF) inhibitors, particularly the monoclonal antibody agents infliximab, adalimumab, and golimumab, and biosimilars to these original drugs. Hence, ADA may cause allergic reactions, loss of drug efficacy, and increased cost of therapy. The risk of developing ADA appears least common with use of etanercept, a TNF receptor fusion protein, and most common with infliximab and adalimumab, a chimeric (mouse/human) antibody construct and a humanized monoclonal antibody, respectively.

The composition and association with ADA induction of each of the four groups of biologic TNF inhibitors that are widely available for the treatment of autoimmune and inflammatory illnesses are:

Chimeric mouse/human anti-TNF antibodies

Infliximab and biosimilars to infliximab – Neutralizing, and to a lesser extent non-neutralizing, anti-infliximab ADA, which are frequently associated with diminished or absent trough levels of the drug and response failure, are a significant concern with the use of infliximab and biosimilars to this agent [1-9]. These drugs are composed of the human constant region of immunoglobulin G1 (IgG1) coupled to the variable regions of a mouse anti-TNF-alpha monoclonal antibody [10]. These ADA are predominantly directed against the mouse portions of the infliximab molecule and are thus sometimes referred to as human antichimeric antibodies (HACA). (See 'Infliximab-induced human antichimeric antibodies' below.)

Human or humanized anti-TNF antibodies

Adalimumab and biosimilars to adalimumab – Adalimumab is composed of a whole human IgG1 kappa monoclonal antibody specific for human TNF-alpha, thus selectively neutralizing TNF-alpha activity [10]. The drug binds to both the soluble and transmembrane bioactive forms of human TNF-alpha. Neutralizing anti-adalimumab ADA pose similar problems to those seen with infliximab, including lower or absent circulating levels of the drug and reduced clinical responsiveness, despite adalimumab being a humanized antibody [7,8,11-14]. The human anti-human antibodies (HAHA) against adalimumab and biosimilars are most likely directed against idiotopes in the TNF-binding sites of the drugs. (See 'Adalimumab-induced human anti-human antibodies' below.)

Golimumab – ADA have also been described in patients receiving golimumab [11,15], a fully human IgG1 kappa monoclonal antibody specific for human TNF-alpha that selectively neutralizes TNF-alpha activity [10]. Adverse clinical consequences of such antibodies, including reduced efficacy, are well described [8,16]. Like infliximab and adalimumab, golimumab binds to both the soluble and transmembrane bioactive forms of human TNF-alpha.

Humanized anti-TNF Fab' fragment

Certolizumab pegol – Certolizumab pegol is a chimeric (mouse/human) anti-TNF Fab' (Fc-free) antibody construct, which consists of the TNF-alpha-binding complementarity-determining region of a monoclonal mouse antibody inserted into a human Fab' IgG framework [10]. Polyethylene glycol is incorporated into the construct, as this may reduce antigenicity and also prolong the circulating half-life of the drug. ADA can also develop against certolizumab pegol [8,11,17]. In one clinical trial including 595 patients with Crohn disease, anti-certolizumab pegol ADA developed in 134 (23 percent) [17]. In 77 percent of the anti-certolizumab pegol-positives, the ADA were persistent and associated with reduced circulating levels of the drug and increased blood levels of C-reactive protein and fecal calprotectin indicating increased disease activity. There are fewer reports of ADA against certolizumab pegol compared with infliximab and adalimumab, and the clinical impact of ADA to this drug is not yet well defined.

TNF-receptor fusion protein

Etanercept and biosimilars to etanercept – Etanercept is a fusion protein consisting of two human p75 TNF-alpha receptors coupled to the constant region of human IgG1 [10]. It binds to and neutralizes both TNF-alpha and TNF-beta. ADA have also been described in patients treated with etanercept, although these are generally non-neutralizing and not associated with decreased trough drug levels, impaired clinical response, or adverse events [11,18]. As methodologic problems in the detection and measurement of anti-etanercept ADA may account for variability among studies, the true immunogenicity of etanercept is still unresolved [18,19].

Biosimilar drugs with pharmacologic, structural, biologic, and therapeutic characteristics that closely match the initially marketed innovator molecules are becoming available in several countries [20-23]. It appears that biosimilars will have similar adverse effects as the original molecules, although larger numbers of patients will have to be treated with such medications before this assumption is confirmed [8,24,25]. The immunogenicity profiles of biosimilars also need to be investigated for each of these drugs, as immunogenicity of an anti-TNF biosimilar drug cannot be deduced solely upon the immunogenicity of the originator drug [26]. Judging by the number of patients with measurable ADA, initial clinical studies of infliximab biosimilars suggest immunogenicity profiles comparable to those of the originator drug. (See "Overview of biologic agents in the rheumatic diseases", section on 'Biosimilars for biologic agents'.)

ADA induced by, and directed against, infliximab and adalimumab appear to be specific to each of these molecules. Cross-reactions with other chimeric monoclonal antibodies such as rituximab have been very rare in our experience.

Clinical effects — The adverse effects of ADA, including increased risk of hypersensitivity reactions, diminished efficacy, and reduced cost-effectiveness, have been documented in a few randomized trials and multiple observational studies [4,8]. Suspicion of ADA development should arise with clinical signs of hypersensitivity reactions and reduced drug efficacy, the latter preferably before intensifying drug delivery as this may increase the risk of side effects and prove to have no or only temporary effect. In drug-naïve patients, serum ADA are usually detectable within two to six months of starting therapy and rarely after 12 months of treatment. A 2017 systematic review of such evidence, which included data from patients treated with infliximab and the biosimilar CT-P13, adalimumab, golimumab, and etanercept, found the following [8]:

Rates of ADA formation were generally higher in patients treated with chimeric anti-TNF constructs (infliximab and biosimilar to infliximab) compared with fully human monoclonal antibodies (golimumab) and the fusion protein etanercept. It is noteworthy that ADA against golimumab seem less likely to occur than ADA against adalimumab, which is developed by phage substitution, possibly making the antigen-binding epitopes immunogenic.

Patients with rheumatoid arthritis, ankylosing spondylitis, psoriasis with and without arthritis, juvenile idiopathic arthritis, Crohn disease, and ulcerative colitis treated with anti-TNF antibody constructs who developed ADA against the drug had lower serum drug levels than ADA-negative patients and were less likely to achieve a significant clinical response at six months and between months 6 and 12.

Discontinuation of the biologic agent in patients with rheumatoid arthritis and/or axial spondyloarthritis with ADA was significantly more likely compared with patients without ADA.

Hypersensitivity reactions among patients with all conditions were more common in ADA-positive patients and most prominent in those treated with infliximab and a biosimilar to infliximab.

Co-therapy with a traditional "nonbiologic" disease-modifying antirheumatic drug (DMARD), methotrexate, azathioprine, leflunomide, or mycophenolate, was associated with decreased measurable levels of circulating ADA. There was no evidence in the literature of any differences between these DMARDs.

Limitations of studies of ADA against drugs that are also antibodies and factors limiting the clinical usefulness of such measurements include the inaccuracy of some assays and the inability of certain assays to determine the in vivo function of ADA (eg, the neutralization capacity of ADA in individual patients) [27-31].

Infliximab-induced human antichimeric antibodies — The development of ADA against infliximab is associated with an increased risk of infusion reactions and a reduced response and duration of response to treatment [1,2,12]. Such ADA have been described in patients with rheumatoid arthritis, inflammatory bowel disease, and other disorders [1-3,6,7,12,22,32-35].

ADA directed against infliximab develop in approximately half of the patients treated with infliximab alone, ie, without other immunosuppressive or antiinflammatory drug therapies. This proportion appears to be reduced by coadministration of methotrexate and other immunoinflammatory modulators [1,33,36,37]. For these reasons, infliximab was approved for use in rheumatoid arthritis in the United States and Europe together with methotrexate. However, such restrictions do not apply to the labeling approved for ankylosing spondylitis, inflammatory bowel disease, and psoriasis, where clinical trials of the effect of methotrexate co-therapy on drug immunogenicity differ considerably. An immunosuppressive action of methotrexate on anti-infliximab ADA development has been proposed, but studies have been limited to measurement of ADA in the circulation and not the ability of methotrexate to interfere with the production of anti-infliximab ADA [27]. An analysis of six randomized trials that examined efficacy, pharmacokinetics, and safety of combination immunomodulator/adalimumab therapies in patients with inflammatory bowel disease failed to show any efficacy benefits of the combination therapies [14].

Examples of the effects of these antibodies in specific conditions include:

Inflammatory bowel disease (Crohn disease and ulcerative colitis) – ADA were common in a cohort of 125 consecutive patients with Crohn disease treated with infliximab (mean of 3.9 infusions [range 1 to 17] over a mean of 10 months) and were associated with an increased risk of infusion reactions and a shorter duration of drug responsiveness [32]. The following observations were made:

Anti-infliximab ADA were detected in 61 percent of patients. The presence of ADA concentrations of 8 mcg/mL or greater before an infusion predicted a shorter mean duration of response compared with patients with lower concentrations (35 versus 71 days).

Higher ADA concentrations (8 mcg/mL or greater) before infusions were associated with a greater risk of an infusion reaction (relative risk 2.4, 95% CI 1.7-3.7), and infliximab concentrations were significantly lower at four weeks among patients who had had an infusion reaction than among patients who had never had an infusion reaction (median 1.2 versus 14.1 mcg/mL). Patients who had infusion reactions had a shorter median duration of clinical response (39 versus 65 days).

The use of concomitant immunosuppressive/antiinflammatory therapy (azathioprine, mercaptopurine, methotrexate, or mesalamine) predicted lower measurable titers of circulating ADA against infliximab and higher concentrations of infliximab four weeks after an infusion.

Rheumatoid arthritis – ADA and a reduction in the duration of the response, which are similar findings to those seen in Crohn disease, have been reported in studies of patients with rheumatoid arthritis [1,6,12,33]. In one study, although low-dose methotrexate administration (7.5 mg/week) appeared to have little effect on the peak clinical response, it prolonged the duration of responses in most patients [33]. In addition, the incidence of ADA positivity correlated inversely with the dose of infliximab (53 versus 21 versus 7 percent of patients who received 1, 3, and 10 mg/kg, respectively). Concomitant methotrexate reduced the frequency of measurable ADA responses to 15, 7, and 0 percent, respectively, in those three treatment groups.

The development of ADA thus has potentially important clinical effects, but uncertainties remain, especially with respect to the clinical utility of assays for the detection of ADA [18,29]. Although ADA have been associated with infusion reactions [32], it is less clear that ADA are responsible for the blunting of clinical responses to infliximab that occur with regular administration over time. In addition, testing for ADA is not widely available, and most assays measure binding of ADA to drug, but not whether this binding has a neutralizing effect in vivo [18,19].

More than half of patients with rheumatoid arthritis receiving infliximab undergo increases in their regular dose over time or shortening of the interval at which the medication is administered [34,35]. In most cases, this "dose creep" correlates with the formation of ADA, but loss of efficacy may also result from the inflammatory disease process no longer being driven by TNF [19]. This seems to be the case in some patients after prolonged TNF inhibition. In addition, the need to increase infliximab doses over time sometimes results from underdosing at the start of therapy. Thus, some patients with rheumatoid arthritis or Crohn disease benefit from infliximab given at doses higher than the recommended 3 mg/kg or more frequently than every eight weeks [35].

Adalimumab-induced human anti-human antibodies — ADA that develop in patients being treated with adalimumab are also termed HAHA. Their presence is associated with lower serum adalimumab concentrations, and findings suggest that the development of ADA contributes to the reduced efficacy of adalimumab monotherapy [12,38]. As an example, the frequency, concentration, and clinical impact of anti-adalimumab ADA were evaluated in 272 consecutive patients with rheumatoid arthritis begun on adalimumab and followed for three years. The following findings were reported:

Anti-adalimumab ADA were detected in 76 patients (28 percent) during 156 weeks of treatment. Antibodies were detected within 28 weeks of beginning therapy in 67 percent of patients who became ADA-positive.

Patients with moderate to high levels of ADA exhibited significantly lower adalimumab levels compared with patients without measurable ADA.

These patients were also significantly less likely to achieve minimal disease activity (Disease Activity Score in 28 Joints [DAS28] <3.2) compared with patients without ADA (6 and 18 versus 48 percent). Additionally, patients with ADA were significantly more likely than those without such antibodies to discontinue study participation due to treatment failure (38 versus 14 percent).

Patients with anti-adalimumab ADA were significantly less likely than those without such antibodies to have also received concomitant therapy with methotrexate (54 versus 82 percent).

CLINICAL IMMUNOGENICITY TESTING — Incorporating immunopharmacologic data regarding circulating drug and anti-drug antibody (ADA) levels (ie, therapeutic drug monitoring [TDM]) has been proposed as a means to rationalize the usual approach to lessening clinical efficacy of infliximab, which is typically either to increase the dose or to increase the frequency with which the medication is administered. The use of TDM and the evidence supporting such use in patients with Crohn disease are described in detail separately. (See "Treatment of Crohn disease in adults: Dosing and monitoring of tumor necrosis factor-alpha inhibitors", section on 'Therapeutic drug monitoring'.)

For drugs under development, regulatory authorities in the United States and Europe now recommend premarketing evaluations for immunogenicity of all novel anti-tumor necrosis factor (TNF) biopharmaceuticals, including biosimilars [26,39]. The use of similar testing in the clinical setting has been proposed by some experts as a means to improve safety and to avoid unnecessary costs of biologic anti-TNF therapies [5,19,32,40,41]. Clinical investigations support these views in patients suffering from rheumatoid arthritis and inflammatory bowel diseases [38,42]. For example, in patients with Crohn disease, proactive and reactive TDM are now endorsed by several gastroenterology associations in the United States [43]. Additionally, a 2019 assembled panel of experts from the United States, Canada, the United Kingdom, and Australia concluded that TDM in patients with Crohn disease and ulcerative colitis is an important tool for optimizing anti-TNF biologic therapies [28]. Limitations to the clinical use of TDM include uncertainties as to when to utilize it in individual patients and evaluations of the optimal window thresholds to target.

AUTOIMMUNITY

Autoantibodies — All of the available tumor necrosis factor (TNF) inhibitors have been associated with an increased risk of developing autoantibodies, including antinuclear antibodies (ANA), which are the most common; anti-double-stranded DNA (anti-dsDNA) antibodies (see 'Systemic lupus erythematosus' below); and antiphospholipid antibodies [37]. The incidence of each varies widely in clinical studies, depending in part on the disease being treated, the drug used, and coadministration of other immune-modulating therapies [37,44-46]. In patients with rheumatoid arthritis, the presence of ANA appears to be a risk factor for the appearance of of anti-drug antibodies (ADA) against infliximab and adalimumab [47].

As examples [37]:

Infliximab use in clinical trials was associated with the development of ANA and anti-dsDNA in 29 to 77 and 10 to 29 percent of patients, respectively.

Etanercept treatment of rheumatoid arthritis patients was associated with ANA and anti-dsDNA antibodies in 11 to 36 and 5 to 15 percent, respectively.

Adalimumab treatment in rheumatoid arthritis patients was associated with the development of ANA and dsDNA antibodies in 13 and 5 percent, respectively.

Anticardiolipin and antinucleosome antibodies have also been reported in a few rheumatoid arthritis patients treated with TNF inhibitors, but have only been anecdotally associated with clinical manifestations. Anticardiolipin antibodies were very infrequent (<5 percent) [44]; antinucleosome antibodies developed more frequently with therapy (7 to 26 percent) and were often associated with ANA positivity [46].

Autoimmune diseases — Some patients treated with the biologic TNF inhibitors develop a clinically evident autoimmune condition, presumably as a result of therapy; however, this occurs only in a small fraction of patients despite the substantial frequency of autoantibodies in patients receiving these drugs [48,49]. The majority of cases of autoimmune disease reported among patients receiving TNF inhibitors have been vasculitic syndromes, most often cutaneous vasculitis; lupus-like syndromes; or psoriatic skin changes. In most of these patients, the condition resolves with discontinuation of the offending drug and use of medications of a different class, but some patients require therapy specifically targeting the complicating autoimmune manifestations [48-51]. It appears that conventional disease-modifying antirheumatic drugs (DMARDs) may lower the risk of infusion reactions and lupus- and vasculitis-like events [52].

Systemic autoimmune conditions associated with TNF inhibitor use include:

Drug-induced lupus syndromes (see 'Systemic lupus erythematosus' below)

Systemic vasculitis (see 'Vasculitis' below)

Sarcoidosis (see "Tumor necrosis factor-alpha inhibitors: An overview of adverse effects", section on 'Pulmonary disease')

Antiphospholipid syndrome [48]

Dermatomyositis (DM)/polymyositis (PM) [53]

Organ-specific autoimmune disorders include:

Cutaneous vasculitis (see 'Vasculitis' below)

Psoriatic skin lesions and other cutaneous reactions (see "Tumor necrosis factor-alpha inhibitors: An overview of adverse effects", section on 'Psoriatic skin lesions' and "Tumor necrosis factor-alpha inhibitors: An overview of adverse effects", section on 'Cutaneous reactions')

Uveitis and other inflammatory eye disease, including optic neuritis (see 'Other conditions' below)

Interstitial lung disease (ILD) (see 'Other conditions' below)

Demyelinating disease, including multiple sclerosis (MS) and MS-like disease, optic neuritis, and peripheral neuropathies (both demyelinating and others [49,54]) (see "Tumor necrosis factor-alpha inhibitors: An overview of adverse effects", section on 'Demyelinating disease')

Autoimmune hepatitis [49,54,55]

Several lines of evidence support the view that TNF inhibitor use is causally linked to these syndromes, particularly vasculitis [48,56], including:

The temporal association between the use of TNF inhibitor therapy and the development of clinical features of an autoimmune disease

The resolution of the autoimmune condition following the cessation of targeted TNF treatment

The original appearance in some cases of leukocytoclastic vasculitis at the site of etanercept injections

The reappearance or worsening of the symptoms upon re-exposure to the TNF inhibitor in a majority of cases in which rechallenge has been attempted [56]

Vasculitis — Most cases of vasculitis are confined to the skin and demonstrate findings of a leukocytoclastic vasculitis, although a minority of patients with vasculitis have systemic involvement, usually of the peripheral nerves or kidney [48].

In a series that analyzed all 233 cases of autoimmune disease induced by TNF inhibitors and reported in the medical literature through December 2006, 113 cases of vasculitis were described [56]. Ninety-eight (87 percent) of the patients with vasculitis had cutaneous involvement. Among these, the following lesions were observed:

Palpable purpura – 55 patients (57 percent)

Erythematous papules/macules/punctate lesions – 11 (11 percent)

Ulcers – 9 (9 percent)

Nodules – 9 (9 percent)

In terms of visceral involvement by vasculitis, the peripheral nerves and kidneys were affected in 18 (16 percent) and 15 (13 percent) patients, respectively.

Eighty-three (73 percent) of the 113 vasculitis cases were confirmed histopathologically. Among these, leukocytoclastic vasculitis was reported in 52 patients (63 percent), necrotizing vasculitis in 14 (17 percent), lymphocytic vasculitis in 5 (6 percent), and other findings in 12 (14 percent).

Ninety-two percent of the cases of vasculitis resolved following the discontinuation of TNF inhibitors. Vasculitis recurred in 12 of 16 (75 percent) patients rechallenged with a TNF inhibitor.

Systemic lupus erythematosus — Most cases of drug-induced lupus associated with the use of a biologic TNF inhibitor are characterized by mucocutaneous features, arthralgia or polyarthritis, and pleuropericardial abnormalities, along with the formation of autoantibodies such as those to dsDNA, the Sm antigen, or histones [56-66]. Renal and neurologic manifestations are extremely rare, and the nature of their association with use of a TNF inhibitor remains uncertain [60]. Estimates of the frequency with which patients satisfy classification criteria for systemic lupus erythematosus (SLE) range from 28 to 64 percent [56,63,66]. The syndrome generally resolves after drug discontinuation, although many of the reported patients have also received glucocorticoid therapy. (See "Drug-induced lupus", section on 'Causative drugs'.)

In a representative series that analyzed all 233 cases of autoimmune disease induced by TNF inhibitors reported in the medical literature through December 2006, 92 cases of SLE or lupus-like illness were described (39 percent) [56]. Among the 72 patients in whom sufficient information was available to determine the frequency of the 1997 updated American College of Rheumatology (ACR) criteria for SLE [67], only 37 (51 percent) met the required threshold of fulfilling four or more criteria. This figure was just 35 percent if features already present before TNF inhibitors were started were excluded from consideration.

The frequencies of the following features were observed:

ANA – 79 percent

Anti-dsDNA antibodies – 72 percent

Cutaneous features – 67 percent

Arthritis – 31 percent

In this series and others, serositis was less common than rash or arthritis, and renal and neurologic symptoms were rare [61,62]. The frequency of antihistone antibodies, a hallmark of drug-induced lupus caused by procainamide or hydralazine, ranged from only 17 to 57 percent [57,61].

The rarity of drug-induced lupus due to TNF inhibitors was illustrated in observations from a large French registry, in which the incidence among patients treated with infliximab or etanercept was approximately 0.2 percent, with too few data for adalimumab to make an accurate estimate [57], although lupus-like syndromes have been reported in some adalimumab-treated patients [56,58].

In one of the largest series, the symptoms and signs of SLE resolved following the discontinuation of the TNF inhibitor in 71 of the 72 patients in whom information regarding disease outcome was available [56]. Resolution of SLE was seen in other series as well, although many of the patients also received glucocorticoids for the related symptoms [61]. The use of concomitant methotrexate, which is thought to reduce the frequency of autoantibodies when using anti-TNF agents, did not appear to protect against the development of TNF inhibitor-induced SLE or lupus-like illness.

Other conditions — Additional conditions with autoimmune features have been described (see 'Autoimmune diseases' above):

Interstitial lung disease – Pulmonary disease, particularly ILD, has been reported in at least 119 patients in association with use of a TNF inhibitor [68,69]. However, the relationship between TNF inhibitor use and the development of ILD is less clear than the association with cutaneous vasculitis or lupus-related syndromes. In most cases in which the lung disease has taken an aggressive course, the patients have had underlying disorders such as rheumatoid arthritis that place them at risk for ILD, making it impossible to exclude confounding by indication as a factor in these reports. In a 2011 literature review, the prognosis in patients with TNF inhibitor-related ILD was poor in patients in whom outcomes were reported, with an overall mortality rate of approximately one-third, and a mortality rate of two-thirds in those with pre-existing ILD exacerbated by use of the drugs [69]. (See "Tumor necrosis factor-alpha inhibitors: An overview of adverse effects", section on 'Pulmonary disease' and "Drug-induced lung disease in rheumatoid arthritis", section on 'Biological agents'.)

Uveitis – Ocular inflammation of the uveal tract (uveitis) has been reported in association with the use of anti-TNF agents. It is most common in patients with ankylosing spondylitis [70]. Notably, the TNF inhibitors are also used to treat uveitis of multiple etiologies. (See "Uveitis: Treatment", section on 'Anti-tumor necrosis factor-alpha'.)

One registry-based study that included spontaneous reports of uveitis during the period from January 1, 1998 through January 1, 2006 noted 43 cases in patients who were receiving anti-TNF therapy [71]. After exclusion of patients with diseases associated with an increased risk of uveitis (eg, ankylosing spondylitis, Crohn disease, or juvenile idiopathic arthritis), the numbers of patients taking etanercept, infliximab, or adalimumab in the remaining cases were 20, 4, and 2, respectively. The authors of this study concluded that, although the risk of uveitis was greater, this should not influence the initial choice of anti-TNF agent, but they did suggest that, for a patient without an underlying disease known to predispose to uveitis who develops uveal inflammation during treatment with etanercept, a switch to an anti-TNF antibody (infliximab or adalimumab) would be an option to consider. Other evidence suggests a preference for the use of monoclonal anti-TNF agents, such as infliximab or adalimumab, rather than etanercept, in patients with ankylosing spondylitis with an active or previous history of uveitis [50,72]. (See "Uveitis: Treatment", section on 'Anti-tumor necrosis factor-alpha'.)

Inflammatory myopathy – At least 20 patients have been described with new onset of DM or PM during treatment with TNF inhibitor therapy, most of whom were being treated for rheumatoid arthritis [53]. Six of the patients exhibited antisynthetase antibodies, most were ANA-positive, and some patients had ILD. Improvement of the DM/PM was reported in most patients with withdrawal of the presumed offending agent and treatment with other immune suppressive therapies.

SUMMARY

The risk of forming neutralizing anti-drug antibodies (ADA), which may result in diminished drug efficacy and increased risk of adverse reactions, varies with the specific type of tumor necrosis factor (TNF) inhibitor. ADA are most commonly observed with use of infliximab, adalimumab, and their respective biosimilars. Neutralizing ADA occur extremely rarely with etanercept. Importantly, most commonly used assays do not reveal whether or not reported ADA are neutralizing in vivo. (See 'Anti-drug antibodies' above.)

Human antichimeric and anti-human ADA formed in patients treated with infliximab and adalimumab, respectively, as well as with their biosimilars, can lead to infusion reactions and local and systemic immune reactions, a reduced duration of response to treatment, requirements for higher infliximab doses or for more frequent dosing intervals, lower cost-effectiveness, and eventually to drug failure. Higher levels of ADA are associated with reduced clinical benefit more often in patients treated with infliximab and adalimumab than with agents associated with lower levels of ADA. (See 'Infliximab-induced human antichimeric antibodies' above and 'Adalimumab-induced human anti-human antibodies' above.)

Coadministration of methotrexate (usually approximately 10 mg weekly) and several other agents is associated with reduced circulating levels of ADA and increased drug levels in some infliximab- and adalimumab-treated patients; however, whether these changes are associated with reduced clinical responsiveness may differ depending upon the underlying disease, being more common in RA but not in patients with inflammatory bowel disease. (See 'Infliximab-induced human antichimeric antibodies' above and 'Adalimumab-induced human anti-human antibodies' above.)

Regulatory authorities in the United States and Europe request premarketing evaluations for immunogenicity (ADA testing) for all novel anti-TNF biologics, including biosimilars. ADA testing of patients on prolonged therapy with anti-TNF biologics may predict future response to therapy. (See 'Clinical immunogenicity testing' above.)

For patients with Crohn disease with treatment failure while receiving infliximab therapy, individualized management (based upon serum infliximab and ADA levels) is more cost-effective than standard intensification of drug dosing and without negative impact on clinical efficacy. (See 'Infliximab-induced human antichimeric antibodies' above.)

The formation of autoantibodies such as antinuclear antibodies (ANA), anti-double-stranded DNA (anti-dsDNA), and antiphospholipid antibodies has been reported in response to all biologic TNF inhibitors. (See 'Autoantibodies' above.)

Cases of vasculitis, particularly cutaneous vasculitis, and lupus-like syndromes have been reported in association with biologic TNF inhibitor use. Most cases of vasculitis and systemic lupus erythematosus (SLE) reported in association with the use of a TNF inhibitor resolve following discontinuation of the TNF inhibitor. (See 'Autoimmune diseases' above and 'Vasculitis' above and 'Systemic lupus erythematosus' above.)

Other autoimmune conditions that may develop in association with the use of anti-TNF antibody constructs include psoriatic skin lesions, sarcoidosis, antiphospholipid syndrome, uveitis, interstitial lung disease (ILD), dermatomyositis (DM) and polymyositis (PM), demyelinating disease, peripheral neuropathies, and autoimmune hepatitis. (See 'Autoimmune diseases' above and 'Other conditions' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges John Stone, MD, who contributed to an earlier version of this topic review.

  1. Bendtzen K, Geborek P, Svenson M, et al. Individualized monitoring of drug bioavailability and immunogenicity in rheumatoid arthritis patients treated with the tumor necrosis factor alpha inhibitor infliximab. Arthritis Rheum 2006; 54:3782.
  2. Wolbink GJ, Vis M, Lems W, et al. Development of antiinfliximab antibodies and relationship to clinical response in patients with rheumatoid arthritis. Arthritis Rheum 2006; 54:711.
  3. Baert F, Noman M, Vermeire S, et al. Influence of immunogenicity on the long-term efficacy of infliximab in Crohn's disease. N Engl J Med 2003; 348:601.
  4. Steenholdt C, Brynskov J, Thomsen OØ, et al. Individualised therapy is more cost-effective than dose intensification in patients with Crohn's disease who lose response to anti-TNF treatment: a randomised, controlled trial. Gut 2014; 63:919.
  5. Schaeverbeke T, Truchetet ME, Kostine M, et al. Immunogenicity of biologic agents in rheumatoid arthritis patients: lessons for clinical practice. Rheumatology (Oxford) 2016; 55:210.
  6. Kelly OB, Donnell SO, Stempak JM, et al. Therapeutic Drug Monitoring to Guide Infliximab Dose Adjustment is Associated with Better Endoscopic Outcomes than Clinical Decision Making Alone in Active Inflammatory Bowel Disease. Inflamm Bowel Dis 2017; 23:1202.
  7. Hindryckx P, Novak G, Vande Casteele N, et al. Incidence, Prevention and Management of Anti-Drug Antibodies Against Therapeutic Antibodies in Inflammatory Bowel Disease: A Practical Overview. Drugs 2017; 77:363.
  8. Strand V, Balsa A, Al-Saleh J, et al. Immunogenicity of Biologics in Chronic Inflammatory Diseases: A Systematic Review. BioDrugs 2017; 31:299.
  9. Goncalves J, Santos M, Acurcio R, et al. Antigenic response to CT-P13 and infliximab originator in inflammatory bowel disease patients shows similar epitope recognition. Aliment Pharmacol Ther 2018; 48:507.
  10. Bendtzen K. Anti-TNF-α biotherapies: perspectives for evidence-based personalized medicine. Immunotherapy 2012; 4:1167.
  11. Vincent FB, Morand EF, Murphy K, et al. Antidrug antibodies (ADAb) to tumour necrosis factor (TNF)-specific neutralising agents in chronic inflammatory diseases: a real issue, a clinical perspective. Ann Rheum Dis 2013; 72:165.
  12. Radstake TR, Svenson M, Eijsbouts AM, et al. Formation of antibodies against infliximab and adalimumab strongly correlates with functional drug levels and clinical responses in rheumatoid arthritis. Ann Rheum Dis 2009; 68:1739.
  13. l'Ami MJ, Kneepkens EL, Nurmohamed MT, et al. Long-term treatment response in rheumatoid arthritis patients starting adalimumab or etanercept with or without concomitant methotrexate. Clin Exp Rheumatol 2017; 35:431.
  14. Colombel JF, Jharap B, Sandborn WJ, et al. Effects of concomitant immunomodulators on the pharmacokinetics, efficacy and safety of adalimumab in patients with Crohn's disease or ulcerative colitis who had failed conventional therapy. Aliment Pharmacol Ther 2017; 45:50.
  15. Vande Casteele N, Khanna R. Therapeutic Drug Monitoring of Golimumab in the Treatment of Ulcerative Colitis. Pharm Res 2017; 34:1556.
  16. Zhou H, Jang H, Fleischmann RM, et al. Pharmacokinetics and safety of golimumab, a fully human anti-TNF-alpha monoclonal antibody, in subjects with rheumatoid arthritis. J Clin Pharmacol 2007; 47:383.
  17. Sandborn WJ, Wolf DC, Kosutic G, et al. Effects of Transient and Persistent Anti-drug Antibodies to Certolizumab Pegol: Longitudinal Data from a 7-Year Study in Crohn's Disease. Inflamm Bowel Dis 2017; 23:1047.
  18. Bendtzen K. Immunogenicity of Anti-TNF-α Biotherapies: II. Clinical Relevance of Methods Used for Anti-Drug Antibody Detection. Front Immunol 2015; 6:109.
  19. Bendtzen K. Personalized medicine: theranostics (therapeutics diagnostics) essential for rational use of tumor necrosis factor-alpha antagonists. Discov Med 2013; 15:201.
  20. Kay J, Feagan BG, Guirguis MS, et al. Health Canada/BIOTECanada Summit on regulatory and clinical topics related to subsequent entry biologics (biosimilars), Ottawa, Canada, 14 May 2012. Biologicals 2012; 40:517.
  21. Minghetti P, Rocco P, Cilurzo F, et al. The regulatory framework of biosimilars in the European Union. Drug Discov Today 2012; 17:63.
  22. Smits LJ, Derikx LA, de Jong DJ, et al. Clinical Outcomes Following a Switch from Remicade® to the Biosimilar CT-P13 in Inflammatory Bowel Disease Patients: A Prospective Observational Cohort Study. J Crohns Colitis 2016; 10:1287.
  23. Benucci M, Gobbi FL, Bandinelli F, et al. Safety, efficacy and immunogenicity of switching from innovator to biosimilar infliximab in patients with spondyloarthritis: a 6-month real-life observational study. Immunol Res 2017; 65:419.
  24. Feagan BG, Choquette D, Ghosh S, et al. The challenge of indication extrapolation for infliximab biosimilars. Biologicals 2014; 42:177.
  25. Braun J, Kudrin A. Progress in biosimilar monoclonal antibody development: the infliximab biosimilar CT-P13 in the treatment of rheumatic diseases. Immunotherapy 2015; 7:73.
  26. https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-immunogenicity-assessment-therapeutic-proteins-revision-1_en.pdf (Accessed on June 08, 2019).
  27. Prado MS, Bendtzen K, Andrade LEC. Biological anti-TNF drugs: immunogenicity underlying treatment failure and adverse events. Expert Opin Drug Metab Toxicol 2017; 13:985.
  28. Papamichael K, Cheifetz AS, Melmed GY, et al. Appropriate Therapeutic Drug Monitoring of Biologic Agents for Patients With Inflammatory Bowel Diseases. Clin Gastroenterol Hepatol 2019; 17:1655.
  29. Atiqi S, Hooijberg F, Loeff FC, et al. Immunogenicity of TNF-Inhibitors. Front Immunol 2020; 11:312.
  30. Mehta P, Manson JJ. What Is the Clinical Relevance of TNF Inhibitor Immunogenicity in the Management of Patients With Rheumatoid Arthritis? Front Immunol 2020; 11:589.
  31. Di Paolo A, Luci G. Personalized Medicine of Monoclonal Antibodies in Inflammatory Bowel Disease: Pharmacogenetics, Therapeutic Drug Monitoring, and Beyond. Front Pharmacol 2020; 11:610806.
  32. Maneiro JR, Salgado E, Gomez-Reino JJ. Immunogenicity of monoclonal antibodies against tumor necrosis factor used in chronic immune-mediated Inflammatory conditions: systematic review and meta-analysis. JAMA Intern Med 2013; 173:1416.
  33. Maini RN, Breedveld FC, Kalden JR, et al. Therapeutic efficacy of multiple intravenous infusions of anti-tumor necrosis factor alpha monoclonal antibody combined with low-dose weekly methotrexate in rheumatoid arthritis. Arthritis Rheum 1998; 41:1552.
  34. Ollendorf DA, Massarotti E, Birbara C, Burgess SM. Frequency, predictors, and economic impact of upward dose adjustment of infliximab in managed care patients with rheumatoid arthritis. J Manag Care Pharm 2005; 11:383.
  35. St Clair EW, Wagner CL, Fasanmade AA, et al. The relationship of serum infliximab concentrations to clinical improvement in rheumatoid arthritis: results from ATTRACT, a multicenter, randomized, double-blind, placebo-controlled trial. Arthritis Rheum 2002; 46:1451.
  36. Elliott MJ, Maini RN, Feldmann M, et al. Repeated therapy with monoclonal antibody to tumour necrosis factor alpha (cA2) in patients with rheumatoid arthritis. Lancet 1994; 344:1125.
  37. Atzeni F, Talotta R, Salaffi F, et al. Immunogenicity and autoimmunity during anti-TNF therapy. Autoimmun Rev 2013; 12:703.
  38. Bodio C, Grossi C, Pregnolato F, et al. Personalized medicine in rheumatoid arthritis: How immunogenicity impacts use of TNF inhibitors. Autoimmun Rev 2020; 19:102509.
  39. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/immunogenicity-testing-therapeutic-protein-products-developing-and-validating-assays-anti-drug (Accessed on June 08, 2019).
  40. Vande Casteele N, Gils A. Pharmacokinetics of anti-TNF monoclonal antibodies in inflammatory bowel disease: Adding value to current practice. J Clin Pharmacol 2015; 55 Suppl 3:S39.
  41. Mitrev N, Vande Casteele N, Seow CH, et al. Review article: consensus statements on therapeutic drug monitoring of anti-tumour necrosis factor therapy in inflammatory bowel diseases. Aliment Pharmacol Ther 2017; 46:1037.
  42. Buhl S, Dorn-Rasmussen M, Brynskov J, et al. Therapeutic thresholds and mechanisms for primary non-response to infliximab in inflammatory bowel disease. Scand J Gastroenterol 2020; 55:884.
  43. Vande Casteele N, Feagan BG, Wolf DC, et al. Therapeutic Drug Monitoring of Tumor Necrosis Factor Antagonists in Crohn Disease: A Theoretical Construct to Apply Pharmacokinetics and Guidelines to Clinical Practice. Inflamm Bowel Dis 2021; 27:1346.
  44. Visvanathan S, Wagner C, Smolen J, et al. IgG and IgM anticardiolipin antibodies following treatment with infliximab plus methotrexate in patients with early rheumatoid arthritis. Arthritis Rheum 2006; 54:2840.
  45. Charles PJ, Smeenk RJ, De Jong J, et al. Assessment of antibodies to double-stranded DNA induced in rheumatoid arthritis patients following treatment with infliximab, a monoclonal antibody to tumor necrosis factor alpha: findings in open-label and randomized placebo-controlled trials. Arthritis Rheum 2000; 43:2383.
  46. Benucci M, Saviola G, Baiardi P, et al. Anti-nucleosome antibodies as prediction factor of development of autoantibodies during therapy with three different TNFalpha blocking agents in rheumatoid arthritis. Clin Rheumatol 2008; 27:91.
  47. Mori A, Saito T, Takahashi M, et al. Presence of anti-nuclear antibodies is a risk factor for the appearance of anti-drug antibodies during infliximab or adalimumab therapy in patients with rheumatoid arthritis. PLoS One 2020; 15:e0243729.
  48. Ramos-Casals M, Roberto-Perez-Alvarez, Diaz-Lagares C, et al. Autoimmune diseases induced by biological agents: a double-edged sword? Autoimmun Rev 2010; 9:188.
  49. Perez-Alvarez R, Pérez-de-Lis M, Ramos-Casals M, BIOGEAS study group. Biologics-induced autoimmune diseases. Curr Opin Rheumatol 2013; 25:56.
  50. Brito-Zerón P, Perez-Alvarez R, Ramos-Casals M, BIOGEAS Study Group. Etanercept and uveitis: friends or foes? Curr Med Res Opin 2015; 31:251.
  51. Boyman O, Comte D, Spertini F. Adverse reactions to biologic agents and their medical management. Nat Rev Rheumatol 2014; 10:612.
  52. Jani M, Dixon WG, Chinoy H. Drug safety and immunogenicity of tumour necrosis factor inhibitors: the story so far. Rheumatology (Oxford) 2018; 57:1896.
  53. Brunasso AM, Aberer W, Massone C. New onset of dermatomyositis/polymyositis during anti-TNF-α therapies: a systematic literature review. ScientificWorldJournal 2014; 2014:179180.
  54. Stübgen JP. Tumor necrosis factor-alpha antagonists and neuropathy. Muscle Nerve 2008; 37:281.
  55. Czaja AJ. Drug-induced autoimmune-like hepatitis. Dig Dis Sci 2011; 56:958.
  56. Ramos-Casals M, Brito-Zerón P, Muñoz S, et al. Autoimmune diseases induced by TNF-targeted therapies: analysis of 233 cases. Medicine (Baltimore) 2007; 86:242.
  57. De Bandt M, Sibilia J, Le Loët X, et al. Systemic lupus erythematosus induced by anti-tumour necrosis factor alpha therapy: a French national survey. Arthritis Res Ther 2005; 7:R545.
  58. Adalimumab (Humira) package insert.
  59. De Bandt M. Lessons for lupus from tumour necrosis factor blockade. Lupus 2006; 15:762.
  60. Stokes MB, Foster K, Markowitz GS, et al. Development of glomerulonephritis during anti-TNF-alpha therapy for rheumatoid arthritis. Nephrol Dial Transplant 2005; 20:1400.
  61. Williams EL, Gadola S, Edwards CJ. Anti-TNF-induced lupus. Rheumatology (Oxford) 2009; 48:716.
  62. Soforo E, Baumgartner M, Francis L, et al. Induction of systemic lupus erythematosus with tumor necrosis factor blockers. J Rheumatol 2010; 37:204.
  63. Wetter DA, Davis MD. Lupus-like syndrome attributable to anti-tumor necrosis factor alpha therapy in 14 patients during an 8-year period at Mayo Clinic. Mayo Clin Proc 2009; 84:979.
  64. Williams VL, Cohen PR. TNF alpha antagonist-induced lupus-like syndrome: report and review of the literature with implications for treatment with alternative TNF alpha antagonists. Int J Dermatol 2011; 50:619.
  65. Beigel F, Schnitzler F, Paul Laubender R, et al. Formation of antinuclear and double-strand DNA antibodies and frequency of lupus-like syndrome in anti-TNF-α antibody-treated patients with inflammatory bowel disease. Inflamm Bowel Dis 2011; 17:91.
  66. Moulis G, Sommet A, Lapeyre-Mestre M, et al. Is the risk of tumour necrosis factor inhibitor-induced lupus or lupus-like syndrome the same with monoclonal antibodies and soluble receptor? A case/non-case study in a nationwide pharmacovigilance database. Rheumatology (Oxford) 2014; 53:1864.
  67. Hochberg MC. Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum 1997; 40:1725.
  68. Ramos-Casals M, Perez-Alvarez R, Perez-de-Lis M, et al. Pulmonary disorders induced by monoclonal antibodies in patients with rheumatologic autoimmune diseases. Am J Med 2011; 124:386.
  69. Perez-Alvarez R, Perez-de-Lis M, Diaz-Lagares C, et al. Interstitial lung disease induced or exacerbated by TNF-targeted therapies: analysis of 122 cases. Semin Arthritis Rheum 2011; 41:256.
  70. Wendling D, Paccou J, Berthelot JM, et al. New onset of uveitis during anti-tumor necrosis factor treatment for rheumatic diseases. Semin Arthritis Rheum 2011; 41:503.
  71. Lim LL, Fraunfelder FW, Rosenbaum JT. Do tumor necrosis factor inhibitors cause uveitis? A registry-based study. Arthritis Rheum 2007; 56:3248.
  72. Levy-Clarke G, Jabs DA, Read RW, et al. Expert panel recommendations for the use of anti-tumor necrosis factor biologic agents in patients with ocular inflammatory disorders. Ophthalmology 2014; 121:785.
Topic 7974 Version 25.0

References

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