Skip to main content
Therapeutic Advances in Musculoskeletal Disease logoLink to Therapeutic Advances in Musculoskeletal Disease
. 2020 Jun 22;12:1759720X20930116. doi: 10.1177/1759720X20930116

Tuberculosis and targeted synthetic or biologic DMARDs, beyond tumor necrosis factor inhibitors

Gerasimos Evangelatos 1,, Vasiliki Koulouri 2, Alexios Iliopoulos 3, George E Fragoulis 4,5
PMCID: PMC7309385  PMID: 32612710

Abstract

Patients with autoimmune rheumatic diseases (ARD) have an increased risk for tuberculosis (TB). The use of tumor necrosis factor inhibitors (TNFi) and glucocorticoids in these patients has been associated with an increased prevalence of latent TB reactivation. Over the last few years, several biologic disease-modifying anti-rheumatic drugs (bDMARDs), other than TNFi (e.g. rituximab, abatacept, tocilizumab, secukinumab) and targeted synthetic DMARDs (tsDMARDs) [e.g. apremilast, Janus kinase (JAK) inhibitors] have been used for the treatment of patients with ARD. For many of these drugs, especially the newer ones like JAK inhibitors or antibodies against interleukin (IL)-23, most data stem from randomized clinical trials and few are available from real life clinical experience. We sought to review the current evidence for TB risk in patients with ARD treated with tsDMARDs or bDMARDs, other than TNFi. It seems that some of these drugs are associated with a lower TB risk, indirectly compared with TNFi treatment. In fact, it appears that rituximab, apremilast and inhibitors of IL-17 and IL-23 might be safer, while more data are needed for JAK inhibitors. As seen in TNFi, risk for TB is more pronounced in TB-endemic areas. Screening for latent TB must precede initiation of any tsDMARDs or bDMARDs. The growing use of non-TNFi agents has raised the need for more real-life studies that would compare the risk for TB between TNFi and other treatment modalities for ARD. Knowledge about the TB-safety profile of these drugs could help in the decision of drug choice in patients with confirmed latent TB infection or in TB endemic areas.

Keywords: autoimmune, biologic DMARDs, rheumatic disease, targeted synthetic DMARDs, tuberculosis, Tumor necrosis factor inhibitor

Introduction

Tuberculosis (TB) has been recognized as an important opportunistic infection occurring in patients with autoimmune rheumatic diseases (ARD). Data from large nationwide registries have shown that the risk of latent TB reactivation or de novo TB cases is increased in ARD patients treated with tumor necrosis factor (TNF) inhibitors (TNFi).110 This seems to be more pronounced in countries that are endemic for TB.6

Mycobacterium tuberculosis (MT) contamination can lead to three possible outcomes: eradication of MT, latent or active MT infection. Ideally, successful eradication of the MT can be achieved by the first line of defence, which comprises alveolar macrophages and other phagocytes. Should innate immunity fail to eliminate the pathogen, active TB develops or the infection is limited through the formation of granulomas, which is mainly mediated by T cells. The tuberculous granuloma consists of macrophages and a surrounding layer of lymphocytes acting protectively for the host. On the other hand, granuloma works as a nest for some MT bacilli that survive inside for long period. This is the stage of latent TB infection. Any factor that leads to immunosuppression might disturb the delicate balance of latent TB and result in active TB infection (TB reactivation).11 Host responses against TB are mediated through an intricate interplay between innate and adaptive immunity, dominated by macrophages and T cells, respectively. Data regarding humoral immunity are ambiguous, with most studies showing a rather negligible role of B cells.12 From a cytokine point-of-view, TNFα and interferon gamma (IFNγ), are essential for the effective intra-cell communication and for granuloma formation.12 Specifically, TNFα is essential in granuloma formation and has been shown to augment phagocytosis of mycobacteria,13 lead ineffective macrophages to apoptosis14,15 and aid in the recruitment of inflammatory cells,16 while IFNγ is vital in preventing TB dissemination, as seen in several cases of defective IFNγ action.1719 Several studies have shown that TNFα neutralization might lead to de novo TB infection or TB reactivation via inhibition of IFNγ-induced phagosomal maturation,20 granuloma destabilization21 and alteration of T cell cytokine production and subpopulation distribution.22,23 A large number of other cytokines have been also implicated in TB immunity, mainly IFNα/β, IL-1, IL-6, IL-12, IL-17 and IL-22.24

It is known that in rheumatoid arthritis (RA) glucocorticoids and methotrexate carry a slightly increased risk of TB infection25,26 while TNFi offer a 4- to 8-fold risk in this population.1,4,6 This risk seems to be decreased over time as more detailed screening with tuberculin skin test (TST) and interferon gamma release assay (IGRA) is applied to patients who are about to commence treatment with biologic drugs.1 Of note, it is widely accepted that this risk is significantly lower for soluble receptor of TNF (etanercept) than with monoclonal antibodies against TNF27,28 (Table 1). This might stem from pharmacokinetic and pharmacodynamic disparities between different TNFi.29 Significantly, some patients treated with TNFi that had a negative baseline TST or IGRA test might develop a positive test during treatment period (seroconversion).30

Table 1.

Cases of tuberculosis (TB) and incidence rate (IR) in patients receiving TNF inhibitors.

TNF Inhibitors
Drug Disease Study type No~ Pt-yrs Active TB cases IR§ Rate general population* Reference
Adalimumab RA, AS, PsA, PsO, CD, UC LTE NA 12,757.7 30 184.79 International Souto et al.27
Certolizumab RA LTE NA 9277.0 44 474.29 International Souto et al.27
Etanercept RA, AS, PsA, PsO LTE NA 7164.8 3 65.01 International Souto et al.27
Golimumab RA, AS, PsA LTE NA 3209.1 4 172.13 International Souto et al.27
Infliximab RA, AS, PsA, PsO, CD, UC LTE NA 4396.2 13 347.70 International Souto et al.27
Adalimumab RA, AS, PsA, PsO, CD, UC RLS NA NA 28 215.0 8.9 (France) Tubach et al.28
Adalimumab RA RLS 1190 NA 1 90.0 8.0 (UK) Dixon et al.9
Adalimumab RA RLS NA 28,751 24 83.3 8.0 (UK) Rutherford et al.31
Certolizumab RA RLS NA 2247 2 88.8 8.0 (UK) Rutherford et al.31
Etanercept RA RLS 2327 NA 0 NA 3.0 (USA) Wolfe et al.8
Etanercept RA RLS 3596 NA 2 50.0 8.0 (UK) Dixon et al.9
Etanercept RA RLS NA 36.663 17 46.3 8.0 (UK) Rutherford et al.31
Etanercept RA RLS NA NA 4 80.0 5.5 (Sweden) Askling et al.2
Etanercept RA RLS 103 73.67 0 NA 66.0 (Korea) Seong et al.6
Etanercept RA, AS, PsA RLS NA NA 0 NA 9.4 (Spain) Gomez-Reino et al.10
Infliximab RA, AS, PsA, PsO, CD, UC RLS NA NA 35 187.5 8.9 (France) Tubach et al.28
Infliximab RA RLS 6460 NA 4 52.5 3.0 (USA) Wolfe et al.8
Infliximab RA RLS 2878 NA 7 150.0 8.0 (UK) Dixon et al.9
Infliximab RA RLS NA 17,670 13 73.4 8.0 (UK) Rutherford et al.31
Infliximab RA RLS NA NA 9 145.0 5.5 (Sweden) Askling et al.2
Infliximab RA RLS 90 78.17 2 2558.0 66.0 (Korea) Seong et al.6
Infliximab RA, AS, PsA RLS NA NA 17 1113.0 9.4 (Spain) Gomez-Reino et al.10
~

Number of patients included in the study.

§

per 100,000 patient-years.

*

IR for TB infection in general population of certain country per 100,000 population.

AS, ankylosing spondylitis; CD, Crohn’s disease; LTE, long-term extension; NA, not applicable; PsA, psoriatic arthritis; PsO, psoriasis; Pt-yrs, patient-years; RA, rheumatoid arthritis; RLS, real-life study; TNF, tumour necrosis factor; UC, ulcerative colitis.

During the last few years, many new therapeutic modalities have been added to a rheumatologist’s arsenal including monoclonal antibodies, anti-cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) [abatacept (ABA)], anti-CD20 [rituximab (RTX)], anti-IL6 receptor [tocilizumab (TCZ), sarilumab], anti-IL-17 (secukinumab, ixekizumab), anti-IL17 receptor (brodalumab), anti-IL-23/IL-12p40 (ustekinumab), anti-IL23p19 (guselkumab) or small molecules like phosphodiesterase 4 inhibitor (apremilast) and, lately, Janus kinase (JAK) inhibitors (tofacitinib, baricitinib, upatacitinib). Other drugs like newer JAK inhibitors (filgotinib, peficitinib), anti-IL-6 (clazakizumab, sirukumab) and other anti-IL23p19 (risankizumab, tildrakizumab) monoclonal antibodies are also in the pipeline for the treatment of patients with ARD. Herein, we aimed to review the current evidence for the TB risk in patients treated with targeted synthetic (apremilast, JAK inhibitors) or biologic disease modifying antirheumatic drugs (DMARDs), other than TNFi, in immune mediated diseases with a focus on inflammatory arthritis.

A literature search using Pubmed and Embase was made. The key words “Apremilast” OR “Tofacitinib” OR “Baricitinib” OR “Upadacitinib” OR “Filgotinib” OR “Peficitinib” OR “Ustekinumab” OR “Guselkumab” OR “Risankizumab” OR “Tildrakizumab” OR “Secukinumab” OR “Ixekizumab” OR “Brodalumab” OR “Tocilizumab” OR “Sirukumab” OR “Clazakizumab” OR “Sirukumab” OR “Abatacept” OR “Rituximab” AND “Tuberculosis” OR “latent TB” OR “latent tuberculosis” OR “TB” were used. Our search review was not limited in RA and articles concerning other immune-mediated diseases were also considered. Randomized clinical trials, their extension studies as well as real-world studies were included. Reference list of the afore-mentioned articles was also reviewed. Case reports, case series and articles not written in English language were excluded. Our search, covered articles published up to 30 August 2019.

Targeted synthetic DMARDs

Apremilast

Apremilast, an oral phosphodiesterase 4 inhibitor, has demonstrated moderate and sustained efficacy in psoriatic arthritis (PsA).32 Interestingly, in lungs of animal models phosphodiesterase-4 inhibitors CC-3052 and CC-11050 reduced local inflammation and improved the antimicrobial efficacy of isoniazid.33,34 During PALACE 1, PALACE 2, PALACE 3 and PALACE 4 studies, a total of 1644 patients were exposed to apremilast at a 20 mg or 30 mg dose twice daily for 24 to 52 weeks. No de novo TB infection or TB reactivation was reported.3538 Patients in PALACE 1 and PALACE 3 did not undergo baseline screening for latent TB. A 4-year extension (a total of 7465 patient-years) pooled analysis from PALACE 1, PALACE 2 and PALACE 3 did not provide specific data for TB infection, but authors concluded that the long-term risk for opportunistic infections is similar with the first year of apremilast administration and is comparable with placebo group.32 In addition, data from 1184 patients with psoriasis treated for 3 years with apremilast 30mg twice daily revealed no de novo TB infection or TB reactivation.39 Physicians should keep in mind that pathophysiology of TB infection might differ between patients with psoriasis and patients with PsA. There is a lack of long-term real-life data, but two observational studies with 202 PsA patients treated for 6 months reported no TB cases.40,41 Collectively, use of apremilast does not appear to be combined with increased risk for TB infection.

JAK inhibitors

JAK inhibitors (also known as Jakinibs) comprise a new class category of DMARDs. These, block the signal mediated through JAK/signal transducers and activators of transcription (STAT) pathway, which is used by many different cytokines and other molecules.42 JAKs have four members, namely JAK1, JAK2, JAK3 and TYK2. Although Jakinibs have been mainly used in inflammatory arthritis, especially in RA, and in haematological malignancies, it seems that they are efficient in a wide spectrum of immune-mediated diseases such as alopecia areata, inflammatory bowel diseases, dermatomyositis and others.42 Many different Jakinibs have been developed with various selectivity for specific JAK members. In general, limited data are available regarding their safety profile because most of them have been recently approved or are in phase-III trials.

Regarding the pathophysiologic link between this class category and TB, it has been hypothesized that blockade of IL-12 or IL-23 (which act through JAK2/TYK2)43,44 might lead to inhibition of IFNγ production by T cells.45 Besides, mutations in IL-12, TYK2 and STAT1 related genes have been found to associate with inherited susceptibility to mycobacterial diseases.43,46

Tofacitinib

Tofacitinib is the most well studied Jakinib inhibiting JAK3 and JAK1 and to a lesser extend JAK2 and TYK2. Tofacitinib has been approved from US Food and Drug Association (FDA) and European Medicines Agency (EMA) for RA and PsA (5 mg twice daily), as well as for ulcerative colitis (UC) (10 mg twice daily). A study examining data from 14 clinical trials [6 phase-III, 6 phase-II and 2 long-term extension (LTE) studies] enrolling 5671 patients followed-up for 12,664 patient-years47 identified 26 cases of TB with the crude incidence rate (IR) being 210 [95% confidence interval (CI); 140 to 300]/100,000 patient-years. Most of the cases were described in patients receiving high dose tofacitinib (i.e. 10 mg, twice daily). Median time from treatment commencement to TB diagnosis was 64 weeks. Patients aged ⩾65 years-old displayed higher IR compared with younger patients in LTE studies, although this was not the case in data obtained from phase-III studies. Glucocorticoid usage did not seem to alter the risk for TB. It is also noteworthy that in more than half of the patients (58%) TB was extrapulmonary47 as seen in patients treated with TNFi regimes.48 Another similarity with the latter population is that cases of TB were more frequent in endemic countries. Of note, from 263 patients diagnosed with latent TB in the phase-III studies and received chemoprophylaxis with isoniazide, none of them developed TB.47 Similarly, a study assessing the long-term safety of tofacitinib in RA, examining data derived from 6194 patients participating in the phase-I, -II and -III as well as LTE studies, showed that the IR for TB was 200 (100–300)/100,000 person years.49 IRs did not differ between dosing schemes (i.e. 5 mg or 10 mg, twice a day) although they were numerically lower for the 5 mg groups.49 In general, although well designed studies are needed so that a safe conclusion can be drawn, it seems that TB risk offered by tofacitinib is similar to that seen in RA patients treated with TNFi.47

Beyond RA, no LTE studies exist for tofacitinib in PsA. Data from the phase-III studies assessing the efficacy and safety of this drug in PsA have not reported any cases of TB.50,51 Similarly, phase-II and phase-III studies for tofacitinib in UC (10 mg twice daily) did not report any TB cases5255 while results from open label, LTE studies are awaited [ClinicalTrials.gov identifier: NCT01470612]. In a recently published study analysing data from the aforementioned studies, including 1157 patients with total exposure of 1612.8 patient-years, no TB cases are reported.54

Baricitinib

Baricitinib is a more selective Jakinib, inhibiting JAK1 and JAK2. Safety data are derived from studies conducted in patients with RA patients, for which baricitinib has been approved. In the largest study so far, examining data from 8 randomized clinical trials (RCTs) and 1 LTE study, 10 cases of TB were recorded. In a total of 3492 patients with median exposure to the drug of 2.1 years, the respective IR was 150/100,000 person-years.56 All cases were reported in regions with a high prevalence of TB. Along the same lines, in a sub-analysis of patients from east Asia, were TB is endemic, the IR for active TB was 230/100,000 patient-years for baricitinib-treated RA patients.57 However, in a sub-analysis examining Japanese patients enrolled in six of the abovementioned studies, no case of TB was recorded.58 Finally, in another open-label, LTE study examining the safety and efficacy of baricitinib for up to 128 weeks with a total exposure of 433.9 patient-years, in patients with RA, no TB cases were recorded.59

Newer JAK inhibitors

Fewer data are available for the newer JAK inhibitors. These include filgotinib, upadacitinib, both selective for JAK1 and peficitinib which has some selectivity for JAK3.

For upadacitinib, in all phase-III studies for RA patients published so far, enrolling all together more than 1500 patients, only 1 case of TB has been noted.6063 Similarly, for filgotinib and peficitinib that was recently approved in Japan for the treatment of RA, no TB cases have been reported in the published phase-IIb and -III studies.6468 LTE studies and real-world data are needed to further characterize the safety profile of the newer generation Jakinibs.

In conclusion, more data, especially from real-world studies, are needed to define whether the risk for TB is comparable with that seen in patients treated with TNFi or other biologic drugs (Table 2),56 TB screening is sine qua non before commencing these drugs. It should be noted that endemic areas are expected to have more cases of TB, as seen with patients treated with TNFi,47,56 and that many TB cases do not present with the classical manifestations (fever, cough, weight loss) but are extrapulmonary.47,49 Several questions remain unanswered. For example, is the TB risk the same across different Jakinibs? And are there any differences between the different indications for which these drugs are used?

Table 2.

Cases of tuberculosis (TB) and incidence rate (IR) in patients receiving targeted synthetic DMARDs.

Targeted synthetic DMARDs
Drug Disease Study type No~ Pt-yrs Active TB cases IR§ Rate general population* Reference
Apremilast PsO RCT, LTE 1184 3671.3 0 0.0 International Crowley et al.39
Apremilast PsA RCT 1644 NA 0 0.0 International Cutolo et al.35; Edwards et al.36; Kavanaugh et al.37; Wells et al.38
Apremilast PsA RLS 202 101.0 0 0.0 7.0 (Italy) Abignano et al.40; Favalli et al.41
Tofacitinib RA RCT, LTE 5671 12,664.0 26 210.0 International Winthrop et al.47
Tofacitinib RA RCT, LTE 6194 19,406.0 36 200.0 International Cohen, et al.49
Tofacitinib PsA RCT 394 NA 0 0.0 International Gladman et al.50
Tofacitinib PsA RCT 316 NA 0 0.0 International Mease et al.51
Tofacitinib UC RCT 1157 1612.8 0 0.0 International Sandborn et al.54
Baricitinib RA RCT, LTE 3492 6636.7 10 150.0 International Smolen et al.56
Baricitinib RA RCT, LTE 740 1294 3 230.0 East Asia Chen et al.57
Baricitinib RA RCT, LTE 540 851.5 0 0.0 14.0 (Japan) Harigai et al.58
Baricitinib RA LTE 201 433.9 0 0.0 International Keystone et al.59
Upatacitinib RA RCT 2022 NA 1 NA International Burmester et al.60; Fleischmann et al.61; Genovese et al.62; Smolen et al.63
Filgotinib RA RCT 1128 NA 0 0.0 International Genovese et al.65; Kavanaugh et al.66; Westhovens et al.68
Peficitinib RA RCT 545 NA 0 0.0 International Genovese et al.64; Kivitz et al.67
~

Number of patients included in the study.

§

per 100,000 patient-years.

*

IR for TB infection in general population of certain country per 100,000 population.

DMARDs, disease-modifying anti-rheumatic drugs; LTE, long-term extension; NA, not applicable; PsA, psoriatic arthritis; PsO, psoriasis; Pt-yrs, patient-years; RA, rheumatoid arthritis; RCT, randomized control trial; RLS, real-life study; UC, ulcerative colitis.

Biologic DMARDs

IL-12 and IL-23 inhibitors

IL-12 and IL-23 contribute in cellular response to TB, especially in the early phase of the infection, by triggering the expression of IFNγ and TNFα.69 As briefly mentioned previously, indirect blockade of either IL-12 or IL-23 might lead to inhibition of IFNγ production by T cells,45 and mutations in IL-12, TYK2 and STAT1 related genes have been associated with susceptibility to mycobacterial infection.43,46 Of note, it has been shown that the ability to control proliferation of Mycobacterium bovis and granuloma formation was not affected in both IL-23p19-deficient mice and in mice treated with a specific anti-IL-23p19 antibody.70

Ustekinumab

Ustekinumab is a monoclonal antibody against the shared p40 subunit of IL-12 and IL-23, approved for the treatment of PsA and plaque psoriasis. The 1-year safety data from pivotal studies PSUMMIT 1 and PSUMMIT 2 revealed no cases of active TB in a total of 705 PsA patients treated with 45 mg or 90 mg ustekinumab.71,72 Moreover, after 2-year follow-up of 598 patients from the same studies no case of active TB was reported.73 A real-life study with 65 PsA patients treated with ustekinumab for 2 years did not report any data on TB and patients with latent TB were excluded.74 In an RCT of ustekinumab for Crohn’s disease (CD), a patient developed de novo active TB ten months after receiving a single intravenous dose of 130 mg.75 The risk for active TB in RCTs for CD was significantly lower in those treated with ustekinumab, than in those treated with golimumab and infliximab (22, 240 and 390 per 100,000 patient-years, respectively).76 Notably, ustekinumab dosage in CD is much higher than that used in inflammatory arthritis and one could speculate that opportunistic infections might be more frequent in the former subgroup. However, data are very limited to lead to a safe conclusion.

A plethora of data supports that ustekinumab does not increase the risk of new TB infection or reactivation of latent TB in patients with psoriasis. During a 4-year follow-up of 1482 psoriasis patients treated with ustekinumab, no TB cases were reported.77 From 167 psoriasis patients with latent TB treated with ustekinumab and isoniazid, no one presented with TB reactivation.78 A study from Taiwan, an intermediate TB burden country, showed no TB reactivation either with or without chemoprophylaxis after an almost 2-year follow-up in 27 psoriasis and PsA patients with latent TB on ustekinumab.79 In the same study, the seroconversion rate was 7.3% with ustekinumab (Table 3), which is lower compared with 14.3% reported with TNFi in the same population.79,80 Although data from psoriasis and CD patients are reassuring about TB risk and ustekinumab, more real-life data are needed for patients with PsA.

Table 3.

Tuberculosis (TB) screening tests seroconversion rates between different biologic DMARDs and cases with seroconversion that developed active TB.

Drug Disease Patients Number of Conversions Active TB Rate (%) Country Reference
Etanercept RA 62 6 0 9.7 Italy Cuomo et al.81
Etanercept RA, JIA, AS 27 4 0 14.8 Italy Cerda et al.82
Adalimumab RA 60 11 0 18.3 Italy Cuomo et al.81
Adalimumab RA, JIA, AS 18 3 1 16.7 Italy Cerda et al.82
Etanercept, Adalimumab PsO 91 13 0 14.3 Taiwan Cheng et al.80
Infliximab RA 11 1 0 9.1 Italy Cuomo et al.81
Infliximab RA, JIA, AS 15 0 0 0 Italy Cerda et al.82
Certolizumab RA 19 1 0 5.3 Italy Cuomo et al.81
Certolizumab RA, JIA, AS 1 0 0 0 Italy Cerda et al.82
Golimumab RA 16 2 0 12.5 Italy Cuomo et al.81
Golimumab RA, JIA, AS 3 0 0 0 Italy Cerda et al.82
Ustekinumab PsO 109 8 0 7.3 Taiwan Hsiao et al.79
Secukinumab PsO 96 1 0 1 Taiwan Wu et al.83
Tocilizumab RA 44 7 0 15.9 Italy Cuomo et al.81
Tocilizumab RA, JIA, AS 13 1 0 7.7 Italy Cerda et al.82
Abatacept RA 37 6 0 16.2 Italy Cuomo et al.81
Abatacept RA, JIA, AS 8 0 0 0 Italy Cerda et al.82
Rituximab RA 43 0 0 0 Taiwan Chen et al.84

AS, ankylosing spondylitis; DMARDs, disease-modifying anti-rheumatic drugs; JIA, juvenile idiopathic arthritis; PsA, psoriatic arthritis; PsO, psoriasis; RA, rheumatoid arthritis.

Guselkumab

After ustekinumab, the targeted anti-IL-23p19 monoclonal antibody guselkumab was approved for the treatment of PsA. Recently, a phase-II study in 100 patients with PsA revealed no cases of active TB during a 1-year follow-up.85 Guselkumab has shown no statistically significant efficacy in RA patients compared with placebo.86 In the latter trial, no case of TB was reported in 110 guselkumab-treated patients (neither with 50 mg nor with 200 mg dose). In addition, in four phase-III clinical trials in 1283 patients with psoriasis, no increased risk for new TB infection or latent TB reactivation was observed during a 1-year follow-up.87

Risankizumab

Risankizumab is a novel anti-IL-23p19 monoclonal antibody approved for the treatment of psoriasis, which is under investigation in PsA and CD and has failed to show efficacy in ankylosing spondylitis (AS). In 185 PsA patients that received risankizumab for 6 months, no active TB cases are reported.88 As for psoriasis, in two phase-III RCTs (ultIMMa-1, ultIMMa-2) including 588 patients, no opportunistic infection was reported.87 In a recently published head-to-head 1-year study between risankizumab and adalimumab in psoriasis, no case of active TB occurred in 301 risankizumab-treated patients.89 No data for TB cases are available for risankizumab use in AS and CD patients.

Tildrakizumab

Another targeted anti-IL-23p19 monoclonal antibody, tildrakizumab, was recently introduced in psoriasis and is under investigation in PsA, AS and non-radiographic axial spondyloarthritis patients. A 6-month phase-IIb study with PsA patients did not present data for TB infection.90 Of note, in two phase-III clinical trials (reSURFACE 1 and reSURFACE 2) and one phase-II a total of more than 1000 psoriasis patients were exposed to tildrakizumab for about 1000 patient-years and the incidence of severe infections was 1.1–1.6 per 100 patient-years, without specific data for TB.91

IL-17 inhibitors

Secukinumab

Secukinumab is a monoclonal antibody that targets IL-17A and has been proved effective in psoriasis, PsA and AS. In TB infection, IL-17 production from neutrophils enhances host immune response,92 while increased IL-17 levels has been found in bronchoalveolar lavage fluid from TB patients.93 Kammüller et al. utilized an in vitro MT microgranuloma model and administered adalimumab and secukinumab. Microgranulomas treated with adalimumab showed characteristics of MT reactivation in contrast to secukinumab-treated microgranulomas, whose results were comparable with untreated or control-treated microgranulomas.94 Thus, it is suggested that secukinumab does not influence MT dormancy and does not lead to experimental TB reactivation.

From a clinical point of view, a recent pooled analysis of 21 clinical trials of secukinumab (15 trials in psoriasis, 3 in PsA and 3 in AS) including 7355 patients with an overall exposure of 16227 patient-years, showed no case of TB reactivation and one case of Mycobacterium avium infection.95 In addition, post-marketing surveillance data were presented in the same study; during 96,054 patient-years, five new TB cases were recorded (5 per 100,000 patient-years), but no reactivation.95 These results are in line with previous report that reviewed safety data from 10 clinical trials of secukinumab only in psoriasis.96 In the latter, in 3430 patients treated for 2725 subject-years, 146 with latent TB were detected and were given anti-TB chemoprophylaxis; no cases of TB reactivation occurred. In a study from Taiwan with 96 secukinumab-treated patients with psoriasis (40% had PsA also), seroconversion was developed in only one patient (~1%) (Table 3) during a mean follow-up of 12.5 months and no case of active TB was reported.83 Conclusively, in vitro studies, clinical trials and post-marketing surveillance data support that patients on secukinumab are at a low risk for TB infection (Table 4).

Table 4.

Cases of tuberculosis (TB) and relative incidence rate (IR) in patients receiving IL-12, IL-23, IL-17 inhibitors.

IL-12, IL-23, IL-17 Inhibitors
Drug Disease Study type No~ Pt-yrs Active TB cases IR§ Rate general population* Reference
Ustekinumab PsA, PsO, CD RCT 5884 4521 1 22.12 International Ghosh et al.76
Ustekinumab PsA RCT 705 NA 0 NA International Ritchlin et al.72
Ustekinumab PsA LTE 615 NA 0 NA International Kavanaugh et al.73
Ustekinumab PsA RLS 65 NA 0 NA 7.0 (Italy) Chimenti et al.74
Ustekinumab CD RCT 1177 NA 1 NA International Feagan et al.75
Ustekinumab PsO LTE 3117 8998 0 0.0 International Lopez-Ferrer et al.77
Guselkumab PsA Phase II 100 NA 0 NA International Deodhar et al.85
Guselkumab RA Phase II 110 NA 0 NA International Smolen et al.86
Guselkumab PsO RCT 1283 NA 0 NA International Crowley et al.87
Rizankizumab PsA RCT 185 NA 0 NA International Mease et al.88
Rizankizumab PsO RCT 588 NA 0 NA International Crowley et al.87
Rizankizumab PsO RCT 301 NA 0 NA International Reich et al.89
Secukinumab AS, PsA, PsO RCT 7355 16,227 0 NA International Deodhar et al.95
Secukinumab AS, PsA, PsO LTE NA 96,054 1 5.0 International Deodhar et al.95
Secukinumab PsO RCT 3430 2725 0 0.0 International van de Kerkhof et al.96
Secukinumab PsO RLS 96 104.5 0 0.0 43.0 (Taiwan) Wu et al.83
Ixekizumab AS RCT 164 NA 0 NA International van der Heijde et al.97
Ixekizumab PsA RCT 1118 1373 0 0.0 International Mease et al.98
Ixekizumab PsO RCT 5370 13,479 0 0 International Romiti et al.99
Brodalumab AS RCT 80 NA 0 NA International Wei et al.100
Brodalumab PsA RCT 168 NA 0 NA International Mease et al.101
~

Number of patients included in the study.

§

per 100,000 patient-years.

*

IR for TB infection in general population of certain country per 100,000 population.

AS, ankylosing spondylitis; CD, Crohn’s disease; LTE, long-term extension; NA, not applicable; PsA, psoriatic arthritis; PsO, psoriasis; Pt-yrs, patient-years; RCT, randomized control trial; RLS, real-life study.

Ixekizumab

Another IL-17A antagonist has been recently introduced in the treatment of PsA and AS. Ixekizumab has demonstrated clinical efficacy and safety in two RCTs in PsA patients (SPIRIT-P1 and SPIRIT-P2). Overall, 1118 patients received ixekizumab and were exposed for 1373.4 patient-years, demonstrating no de novo TB infection or latent TB reactivation (only two patients had positive IGRA at baseline).98 In the COAST-V clinical trial, 164 patients with AS received ixekizumab for 16 weeks and no active or latent TB was reported.97 Notably, the follow-up time in this study was too short and patients with proved or suspected active or latent TB were excluded at the screening. As with other newer bDMARDs, much more safety data is available from clinical trials in patients with psoriasis. An integrated safety data analysis from 11 clinical studies of ixekizumab in psoriasis accounted for 5730 patients exposed for 13479 patient-years.99 During treatment with ixekizumab, 72 patients (1.3%) developed treatment-emergent latent TB or positive IGRA/TST results, but no cases of active TB.

Brodalumab

An IL-17 receptor-A inhibitor, brodalumab, has been approved for psoriasis and has been shown to be efficacious in PsA and AS. In 168 PsA patients treated for 9–12 months with brodalumab, no case of active TB was reported, although patients with latent TB were excluded if they did not receive prophylactic anti-TB treatment.101 Moreover, in a 16-week phase-III study of brodalumab in 80 AS patients, no data for TB are available, but the serious adverse events rate was comparable with the placebo group.100 The experience of brodalumab in psoriasis is richer but data on TB infection from three 1-year phase-III RCTs (AMAGINE-1, AMAGINE-2 and AMAGINE-3) are not available.102 To be mentioned, patients with a known history of past TB infection or positive screening for TB that did not receive prophylactic anti-TB treatment were excluded from the aforementioned studies. Eventually, data regarding active TB cases in brodalumab-treated patients are inadequate to come to a safe conclusion.

IL-6 inhibitors

Tocilizumab

TCZ is a monoclonal antibody directed against both soluble and membrane IL-6 receptor. It has been approved by the FDA and EMA for the treatment of RA, systemic or polyarticular juvenile inflammatory arthritis (JIA) and refractory giant cell arteritis (GCA). IL-6 appears to play an important protective role against MT, mainly in cases of exposure to high mycobacterial load.103,104 Nevertheless, Ogata et al. showed that TCZ did not hinder IFN-γ production induced by two different MT antigens, contrary to etanercept and infliximab, both of which led to IFN-γ level reduction.105 The minimal influence of TCZ in IFN-γ production suggests that TCZ-treated patients might not demonstrate false negative IGRA (as TNFi-treated patients) and are at a low risk of latent TB infection reactivation.

A thorough systematic review examined the safety of non-TNFi biologic agents (TCZ, RTX, ABA, ustekinumab and secukinumab) by collecting data from RCTs, their respective extended-label and open-label studies and national registries.106 In 15,485 RA patients treated with TCZ no cases of TB were reported, regardless the use of conventional DMARDs (cDMARDs).106 Similar results were described in previous systematic reviews and meta-analyses; in an earlier report by Cantini et al., which also included 4 JIA clinical trials, no active TB cases were disclosed.107 Of note, in GCA patients, two clinical studies have shown TCZ efficacy but did not clarify if patients with latent TB were included and if any patient developed active TB during the study period.108,109 Another previous meta-analysis of RCTs and LTEs enrolling patients with RA, PsA, AS, psoriasis, UC or CD, confirmed the absence of TB associated with TCZ in RCTs of RA patients.27 However, in the LTEs in RA patients, there were 9 cases of TB in 12,905.2 patient-years (70/100,000 patient-years) in the TCZ group, though the estimated pooled IR for TB was still considerably lower compared with TNFi.27

Data from real-world studies in Japan (REAL registry, n = 302), Finland (patients with JIA, n = 6), India (n = 13) and a TB endemic area in the United Kingdom (n = 17) showed no cases of active TB in patients under TCZ therapy.110113 Two real-life studies from Taiwan, one 15-year retrospective and one 3-year prospective, reported no cases of active TB among 31 and 114 patients, respectively.114,115 Similarly, a Brazilian retrospective cohort with 336 patient-years follow-up displayed no cases of TB in RA patients treated with TCZ.116 A big cohort of TCZ-treated patients (n = 16,074) from USA that used claims data did not present data for TB.117

Rutherford et al. analysed data from the British Society for Rheumatology Biologics Register for Rheumatoid Arthritis (BSRBR-RA) in order to reveal the incidence of opportunistic infections in RA patients receiving biologic agents. In the TCZ group there was only one case of TB in 2171 patients, with an IR of 26 per 100,000 patient-years.31 Lastly, a real-world study from Malaysia, a country with a high TB incidence, included a total of 68 courses of TCZ treatment in RA patients and demonstrated 3 cases of TB development,118 and a post-marketing safety report of TCZ in RA patients in Japan disclosed a IR of 220 per 100,000 patient-years for active TB infection.119

Two studies from Italy investigated the conversion rate of TB screening tests in patients under TCZ treatment. Among 44 patients with negative initial TST or IGRA test, treatment with TCZ resulted in seroconversion in seven patients, during a mean period of 24 months (Table 3). Nevertheless, none of the patients developed active TB infection.81 In another study, only one out of 13 patients under TCZ treatment experienced TST conversion and received isoniazide as chemoprophylaxis.82

To conclude, TCZ does not seem to be linked with a significantly increased risk for TB infection, especially in countries with a low burden of TB (Table 5).

Table 5.

Cases of tuberculosis (TB) in patients receiving IL-6 Inhibitors, abatacept, rituximab.

IL-6 Inhibitors, abatacept, rituximab
Drug Disease Study type No~ Pt-yrs Active TB cases Rate§ Rate general population* Reference
Tocilizumab RA RCT, LTE 15,485 NA 0 0 International Cantini et al.106
Tocilizumab JIA RCT 205 NA 0 0 International Cantini et al.107
Tocilizumab RA RCT 3354 NA 0 0 International Souto et al.27
Tocilizumab RA LTE NA 12,905.2 9 75.6 International Souto et al.27
Tocilizumab RA RLS 302 NA 0 0 14 (Japan) Sakai et al.113
Tocilizumab JIA RLS 4 6.4 0 0 4.7 (Finland) Tarkiainen et al.111
Tocilizumab Various ARD RLS 16 NA 0 0 199 (India) Shobha et al.112
Tocilizumab RA, JIA RLS 17 NA 0 0 50 (UK-endemic region) Nisar et al.110
Tocilizumab RA RLS 31 55.49 0 0 43 (Taiwan) Lim et al.114
Tocilizumab RA RLS 114 141.38 0 0 43 (Taiwan) Lin et al.115
Tocilizumab, Abatacept, Rituximab RA RLS 195 NA 0 0 45 (Brazil) Yonekura et al.116
Tocilizumab RA RLS 2171 3861 1 26 8 (UK) Rutherford et al.31
Tocilizumab RA RLS 68 NA 3 NA 92 (Malaysia) Tan et al.118
Tocilizumab RA RLS 3881 1793.5 4 223 14 (Japan) Koike et al.119
Sarilumab RA RCT 1348 NA 0 0 International Lee et al.120
Sarilumab AS RCT 251 NA 0 0 International Sieper et al.121
Clazakizumab RA RCT 298 NA 2 NA International Weinblatt et al.122
Clazakizumab PsA RCT 124 NA 0 0 International Mease et al.123
Sirukumab RA RCT 2193 NA 1 NA International Aletaha et al.124; Takeuchi et al.125,126; Taylor et al.127
Abatacept RA RCT, LTE 8539 NA 1 NA International Cantini et al.106
Abatacept RA RCT 433 433 1 230 International Souto et al.27
Abatacept JIA, PsA, SLE RCT 535 NA 0 0 International Cantini et al.107
Abatacept RA RCT, LTE 4149 12,132 8 70 International Weinblatt et al.128
Abatacept RA, JIA, PsA RLS 1292 NA 0 0 International Nisar et al.110; Tarkiainen et al.111; Shobha et al.112; Lim et al.114; Takahashi et al129; Salmon et al.130
Rituximab Various ARD RCT 5233 NA 0 0 International Cantini et al.107
Rituximab RA RCT, LTE 3194 11,962 2 18 International Souto et al.27
Rituximab RA RLS 5072 17,154 2 12 8 (UK) Rutherford et al.31
Rituximab RA RLS 39 NA 2 NA 92 (Malaysia) Tan et al.118
Rituximab Various ARD RLS 33 NA 0 0 50 (UK-endemic region) Nisar et al.110
Rituximab Various ARD RLS 42 NA 0 0 199 (India) Shobha et al.112
Rituximab JIA RLS 9 8 0 0 4,7 (Finland) Tarkiainen et al.111
Rituximab RA RLS 1303 1629 0 0 9.2 (France) Gottenberg et al.131
Rituximab Various ARD RLS 370 299 0 0 7.3 (Germany) Tony et al.132
Rituximab RA RLS 32 NA 0 0 7.3 (Germany) Xanthouli et al.133
Rituximab RA RLS 2484 NA 1 NA 7.3 (Germany) Wendler et al.134
Rituximab RA RLS 763 6179 2 32 43 (Taiwan) Liao et al.135
~

Number of patients included in the study.

§

per 100,000 patient-years.

*

IR for TB infection in general population of certain country per 100,000 population.

ARD, autoimmune rheumatic diseases; AS, ankylosing spondylitis; JIA, juvenile idiopathic arthritis; LTE, long-term extension; NA, not applicable; PsA, psoriatic arthritis; Pt-yrs, patient-years; RA, rheumatoid arthritis; RCT, randomized control trial; RLS, real-life study; SLE, systematic lupus erythematosus; TCZ, tocilizumab.

Newer IL-6 inhibitors

Sarilumab, a fully human monoclonal antibody against IL-6 receptor, has been approved by the FDA and EMA for RA. None of 1348 RA patients treated with sarilumab for at least 1 year experienced active TB.120 Of note, in a 4-month phase-II study in 301 AS patients, sarilumab was not effective, but no cases of TB were reported.121

Clazakizumab is a monoclonal antibody that targets IL-6 with high affinity and specificity. Two cases of pulmonary TB were reported during a 6-month phase-III RCT of clazakizumab in 298 RA patients (1340/100,000 patient-years), both of which were in TB endemic countries.122 On the other hand, no case of TB occurred in 124 clazakizumab-treated PsA patients after a 6-month follow-up.123

Sirukumab selectively binds to IL-6 and has been investigated in RA. Four phase-III RCTs with a follow-up of 52 weeks included 2193 RA patients and one TB case was reported (46/100,000 patient-years), although in two of these studies patients with past history of TB or ‘chronic or recurrent infections’ were excluded.124127

LTE studies and real-life data are still needed to evaluate the TB risk of the newer IL-6 inhibitors.

Abatacept and rituximab

Abatacept

ABA is a fusion protein consisting of the Fc fragment of IgG1 immunoglobulin and the extracellular domain of CTLA-4 that hinders the stimulation of T cells by binding to the co-stimulatory CD80 and CD86 molecules of antigen presenting cells. It has been approved by the FDA and EMA for the treatment of active RA, JIA and adult PsA. A study in animal models has shown that the use of ABA in mice with chronic TB infection did not result in exacerbation of the TB infection, contrary to the use of anti-murine TNF antibody.136

Only one case of probable active TB was reported in 17 trials of ABA use in 8539 RA patients,106 while none were disclosed among 190 JIA and 128 PsA patients.107 In a meta-analysis of LTEs, ABA displayed a low estimated pooled IR for TB (60/100,000 patient-years).27 Integrated analyses of intravenous short-term and cumulative use of ABA in 8 clinical trials showed that TB occurred in only eight patients (66/100,000 patient-years) and presented after 1–3 years of treatment.128

To our knowledge, there have been no cases of active TB reported in several studies conducted in real-life settings in patients with inflammatory arthritis treated with ABA, among 1272 patients.110112,114,116,129,130 TST or IGRA conversion occurred in 6 out of 45 patients (13.3%) under ABA; nevertheless, none of them developed active TB.81,82 In conclusion, it seems that ABA does not significantly increase the risk for de novo or reactivated TB infection.

Rituximab

RTX is a monoclonal antibody targeted against the CD20 protein found on B lymphocytes. This regime is marketed for RA, granulomatosis with polyangiitis (GPA) and microscopic polyangiitis (MPA). As an anti-B cell agent, it does not inhibit T cell action, which is the primary immune cell involved in the protection against TB. An interesting study examined the effects of RTX therapy on IFN-γ levels in 56 patients with RA. Among them, seven patients had latent TB and 6 had TB associated with TNFi treatment. RTX therapy resulted in no significant changes in IFN-γ levels or IGRA conversion and no active TB cases were reported.84

No cases of active TB have been reported in patients receiving RTX in 9 RCTs with RA patients (n = 3623) or patients with Sjogren’s syndrome (n = 107), systematic lupus erythematosus (SLE) (n = 700), mixed cryoglobulinemia (n = 381) and GPA and MPA (n = 422).107 In two LTEs in RA patients, two cases of active TB have been reported during a follow-up time of 9.5 years (18/100,000 patient-years).137 In comparison with other biologics, RTX exhibited the lowest pooled IR of TB in a meta-analysis of LTEs (18 per 100,000 patient-years).27

Data from national registries and real-world data, including patients with several autoimmune conditions other than RA (JIA, SLE, GPA, MPA, multiple sclerosis, pemphigus and other), have confirmed the low risk of de novo TB infection or latent TB reactivation from RTX treatment as no cases of TB have been reported in most of these,110,111,116,131133 even in the presence of latent TB. Only one case of TB was reported in 2484 patients treated with RTX in the German GENIRIS study,134 while two cases with active TB were found in a retrospective Taiwanese RA study during 6179 patient-years (32/100,000 patient-years), both in patients previously treated with TNFi.135 Likewise, data from the BSRBR-RA showed that in the RTX group, only two cases during 17,154 patient-years developed TB (12/100,000 patient-years), which is significantly lower compared with the risk in the TNFi group (65/100,000 patient-years).31

In general, it appears that the risk of TB associated with RTX is lower compared with TNFi, but also to most other bDMARDs and tsDMARDs.27

Conclusion

Based mainly on the results of RCTs and LTE studies, the TB risk associated with the use of most of the non-TNFi agents is generally lower, compared with TNFi. In fact, it seems that the risk of either de novo TB infection or reactivation of latent TB is relatively low with apremilast, ustekinumab, secukinumab and rituximab treatment (Table 6). No safe conclusion can be drawn for Jakinibs yet, but the risk for active TB infection seems comparable with those of TNFi. “Although seroconversion is noted in a small proportion of patients treated with bDMARDs, being lower for those treated with secukinumab and rituximab, this does not lead in increased incidence of latent TB reactivation.”

Table 6.

Comparative presentation of active tuberculosis (TB) incidence rates (IR) between different biologic and targeted synthetic DMARDs.

Drug Disease Study type IR§ Reference
Infliximab RA, AS, PsA, PsO, CD, UC LTE, RLS 52.5–2558.0 Askling et al.2; Seong et al.6; Wolfe et al.8; Dixon et al.9; Gomez-Reino et al.10; Souto et al.27; Tubach et al.28
Certolizumab RA LTE 474.29 Souto et al.27
Adalimumab RA, AS, PsA, PsO, CD, UC LTE, RLS 90.0–215.0 Dixon et al.9; Souto et al.27; Tubach et al.28
Golimumab RA, AS, PsA LTE 172.13 Souto et al.27
Etanercept RA, AS, PsA, PsO RLS, LTE 9.3–80.0 Askling et al.2; Dixon et al.9; Souto et al.27; Tubach et al.28
Rituximab RA RCT, LTE, RLS 0.0–32.0 Rutherford et al.31; van Vollenhoven et al.137; Gottenberg et al.131; Tony et al.132; Xanthouli et al.133; Liao et al.135
Abatacept RA, JIA, PsA RCT, LTE, RLS 0.0–230.0 Souto et al.27; Cantini et al.106,103; Nisar et al.110; Tarkiainen et al.111; Shobha et al.112; Lim et al.114; Weinblatt et al.128; Takahashi et al.129; Salmon et al.130
Tocilizumab RA, JIA RCT, LTE, RLS 0.0–230.0 Souto et al.27; Cantini et al.106; Sakai et al113; Lim et al.114; Lin et al.115; Yonekura et al.116; Rutherford et al.31; Koike et al.119
Apremilast PsA, PsO RCT, LTE, RLS 0.0 Cutolo et al.35; Edwards et al.36; Kavanaugh et al.37; Wells et al.38; Crowley et al.39; Abignano et al.40; Favalli et al.41
Tofacitinib RA RCT, LTE 200.0–210.0 Winthrop et al.47; Cohen et al.49
Baricitinib RA RCT, LTE 150.0–230.0 Smolen et al.56; Chen et al.57
Ustekinumab PsA, PsO, CD RCT, LTE, RLS 0.0–22.12 Ghosh et al.76; Lopez-Ferrer et al.77; Tsai et al.78; Hsiao et al.79
Secukinumab AS, PsA, PsO RCT, LTE 0.0–5.0 Deodhar et al.95; van de Kerkhof et al.96
Ixekizumab PsA, PsO RCT 0.0 Mease et al.98; Romiti et al.99
§

per 100,000 patient-years.

AS, ankylosing spondylitis; CD, Crohn’s disease; DMARDs, disease-modifying anti-rheumatic drugs; JIA, juvenile idiopathic arthritis; LTE, long-term extension; PsA, psoriatic arthritis; PsO, psoriasis; RA, rheumatoid arthritis; RCT, randomized control trial; RLS, real-life study; UC, ulcerative colitis.

In everyday clinical practice, and bearing in mind that new aspects might be enlightened by long-term post-market surveillance, one could say that when there is latent TB, anti-CD20 therapy would be preferable. In diseases where their role is limited (e.g. PsA, seronegative spondyloarthropathies), drugs targeting cytokines involved in the IL-23/-17 axis or apremilast are the most reasonable options. Jakinibs seem to have the same safety profile, regarding TB, with TNFi.

Risk of TB cannot be definitely assessed by data obtained from RCTs.27 RCTs generally tend to underestimate the true incidence of latent TB reactivation, possibly due to the strict patient inclusion criteria and the relatively limited observation time. LTE studies or even better, real-world data from big nation-wide registries are thought to be more appropriate to answer this question.9 The latter are still limited for the most non-TNFi bDMARDs and tsDMARDs. Although TB has been well recognized as an opportunistic infection in the context of RA treated with biologic drugs, it is possibly underestimated in patients receiving non-biologic drugs25 and in other ARD (e.g. SLE). Moreover, when rheumatologists assess the risk for TB infection, they should take into account concomitant cDMARDs or glucocorticoid usage. Finally, it seems that the risk for TB in biologic-exposed patients has been significantly decreased over the last few years.1,31 This is probably related to the increased awareness and subsequent screening. Thus, comparison between studies needs to be interpreted with caution.

Studies specifically designed for assessment of opportunistic infections, including TB, are needed to help the clinician safely use the available drugs, especially in countries endemic for TB and for patients diagnosed with latent TB. Screening for latent TB must always precede bDMARD or tsDMARD initiation.

Footnotes

Conflict of interest: The authors declare that there is no conflict of interest.

Funding: The authors received no financial support for the research, authorship, and/or publication of this article.

ORCID iD: George E Fragoulis Inline graphic https://orcid.org/0000-0003-4932-7023

Contributor Information

Gerasimos Evangelatos, Rheumatology Department, 417 Army Share Fund Hospital (NIMTS), Monis Petraki 10-12, Athens, 11521, Greece.

Vasiliki Koulouri, Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.

Alexios Iliopoulos, Rheumatology Department, 417 Army Share Fund Hospital (NIMTS), Athens, Greece.

George E Fragoulis, Rheumatology Unit, First Department of Propaedeutic Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.

References

  • 1. Arkema EV, Jonsson J, Baecklund E, et al. Are patients with rheumatoid arthritis still at an increased risk of tuberculosis and what is the role of biological treatments? Ann Rheum Dis 2015; 74: 1212–1217. [DOI] [PubMed] [Google Scholar]
  • 2. Askling J, Fored CM, Brandt L, et al. Risk and case characteristics of tuberculosis in rheumatoid arthritis associated with tumor necrosis factor antagonists in Sweden. Arthritis Rheum 2005; 52: 1986–1992. [DOI] [PubMed] [Google Scholar]
  • 3. Brassard P, Lowe AM, Bernatsky S, et al. Rheumatoid arthritis, its treatments, and the risk of tuberculosis in Quebec, Canada. Arthritis Rheum 2009; 61: 300–304. [DOI] [PubMed] [Google Scholar]
  • 4. Carmona L, Hernandez-Garcia C, Vadillo C, et al. Increased risk of tuberculosis in patients with rheumatoid arthritis. J Rheumatol 2003; 30: 1436–1439. [PubMed] [Google Scholar]
  • 5. Gardam M, Iverson K. Rheumatoid arthritis and tuberculosis: time to take notice. J Rheumatol 2003; 30: 1397–1399. [PubMed] [Google Scholar]
  • 6. Seong SS, Choi CB, Woo JH, et al. Incidence of tuberculosis in Korean patients with rheumatoid arthritis (RA): effects of RA itself and of tumor necrosis factor blockers. J Rheumatol 2007; 34: 706–711. [PubMed] [Google Scholar]
  • 7. Yamada T, Nakajima A, Inoue E, et al. Increased risk of tuberculosis in patients with rheumatoid arthritis in Japan. Ann Rheum Dis 2006; 65: 1661–1663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Wolfe F, Michaud K, Anderson J, et al. Tuberculosis infection in patients with rheumatoid arthritis and the effect of infliximab therapy. Arthritis Rheum 2004; 50: 372–379. [DOI] [PubMed] [Google Scholar]
  • 9. Dixon WG, Watson K, Lunt M, et al. Rates of serious infection, including site-specific and bacterial intracellular infection, in rheumatoid arthritis patients receiving anti-tumor necrosis factor therapy: results from the British society for rheumatology biologics register. Arthritis Rheum 2006; 54: 2368–2376. [DOI] [PubMed] [Google Scholar]
  • 10. Gomez-Reino JJ, Carmona L, Valverde VR, et al. Treatment of rheumatoid arthritis with tumor necrosis factor inhibitors may predispose to significant increase in tuberculosis risk: a multicenter active-surveillance report. Arthritis Rheum 2003; 48: 2122–2127. [DOI] [PubMed] [Google Scholar]
  • 11. de Martino M, Galli L, Chiappini E. Reflections on the immunology of tuberculosis: will we ever unravel the skein? BMC Infect Dis 2014; 14(Suppl. 1): S1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. de Martino M, Lodi L, Galli L, et al. Immune response to mycobacterium tuberculosis: a narrative review. Front Pediatr 2019; 7: 350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Bekker LG, Freeman S, Murray PJ, et al. TNF-alpha controls intracellular mycobacterial growth by both inducible nitric oxide synthase-dependent and inducible nitric oxide synthase-independent pathways. J Immunol 2001; 166: 6728–6734. [DOI] [PubMed] [Google Scholar]
  • 14. Keane J, Remold HG, Kornfeld H. Virulent mycobacterium tuberculosis strains evade apoptosis of infected alveolar macrophages. J Immunol 2000; 164: 2016–2020. [DOI] [PubMed] [Google Scholar]
  • 15. Fratazzi C, Arbeit RD, Carini C, et al. Macrophage apoptosis in mycobacterial infections. J Leukoc Biol 1999; 66: 763–764. [DOI] [PubMed] [Google Scholar]
  • 16. Roach DR, Bean AG, Demangel C, et al. TNF regulates chemokine induction essential for cell recruitment, granuloma formation, and clearance of mycobacterial infection. J Immunol 2002; 168: 4620–4627. [DOI] [PubMed] [Google Scholar]
  • 17. Cooper AM, Dalton DK, Stewart TA, et al. Disseminated tuberculosis in interferon gamma gene-disrupted mice. J Exp Med 1993; 178: 2243–2247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Jouanguy E, Lamhamedi-Cherradi S, Altare F, et al. Partial interferon-gamma receptor 1 deficiency in a child with tuberculoid bacillus Calmette-Guerin infection and a sibling with clinical tuberculosis. J Clin Invest 1997; 100: 2658–2664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Flynn JL, Chan J, Triebold KJ, et al. An essential role for interferon gamma in resistance to mycobacterium tuberculosis infection. J Exp Med 1993; 178: 2249–2254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Harris J, Hope JC, Keane J. Tumor necrosis factor blockers influence macrophage responses to mycobacterium tuberculosis. J Infect Dis 2008; 198: 1842–1850. [DOI] [PubMed] [Google Scholar]
  • 21. Chakravarty SD, Zhu G, Tsai MC, et al. Tumor necrosis factor blockade in chronic murine tuberculosis enhances granulomatous inflammation and disorganizes granulomas in the lungs. Infect Immun 2008; 76: 916–926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Hamdi H, Mariette X, Godot V, et al. Inhibition of anti-tuberculosis T-lymphocyte function with tumour necrosis factor antagonists. Arthritis Res Ther 2006; 8: R114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Saliu OY, Sofer C, Stein DS, et al. Tumor-necrosis-factor blockers: differential effects on mycobacterial immunity. J Infect Dis 2006; 194: 486–492. [DOI] [PubMed] [Google Scholar]
  • 24. Domingo-Gonzalez R, Prince O, Cooper A, et al. Cytokines and chemokines in mycobacterium tuberculosis infection. Microbiol Spectr 2016; 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Fragoulis GE, Constantinou CA, Sipsas NV, et al. Tuberculosis in inflammatory arthritis. Are biologics the only culprits? Lancet Rheumatol 2019. (Accepted for publication). [DOI] [PubMed] [Google Scholar]
  • 26. Cantini F, Niccoli L, Capone A, et al. Risk of tuberculosis reactivation associated with traditional disease modifying anti-rheumatic drugs and non-anti-tumor necrosis factor biologics in patients with rheumatic disorders and suggestion for clinical practice. Expert Opin Drug Saf 2019; 18: 415–425. [DOI] [PubMed] [Google Scholar]
  • 27. Souto A, Maneiro JR, Salgado E, et al. Risk of tuberculosis in patients with chronic immune-mediated inflammatory diseases treated with biologics and tofacitinib: a systematic review and meta-analysis of randomized controlled trials and long-term extension studies. Rheumatology (Oxford) 2014; 53: 1872–1885. [DOI] [PubMed] [Google Scholar]
  • 28. Tubach F, Salmon D, Ravaud P, et al. Risk of tuberculosis is higher with anti-tumor necrosis factor monoclonal antibody therapy than with soluble tumor necrosis factor receptor therapy: the three-year prospective French research axed on tolerance of biotherapies registry. Arthritis Rheum 2009; 60: 1884–1894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Ehlers S. Role of tumour necrosis factor (TNF) in host defence against tuberculosis: implications for immunotherapies targeting TNF. Ann Rheum Dis 2003; 62(Suppl. 2): ii37–ii42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Goel N, Torralba K, Downey C, et al. Screening for acquired latent tuberculosis in rheumatoid arthritis patients on tumor necrosis factor inhibition therapy in Southern California. Clin Rheumatol. Epub ahead of print 28 February 2020. DOI: 10.1007/s10067-020-04991-y. [DOI] [PubMed] [Google Scholar]
  • 31. Rutherford AI, Patarata E, Subesinghe S, et al. Opportunistic infections in rheumatoid arthritis patients exposed to biologic therapy: results from the British society for rheumatology biologics register for rheumatoid arthritis. Rheumatology (Oxford) 2018; 57: 997–1001. [DOI] [PubMed] [Google Scholar]
  • 32. Kavanaugh A, Gladman DD, Edwards CJ, et al. Long-term experience with apremilast in patients with psoriatic arthritis: 5-year results from a PALACE 1-3 pooled analysis. Arthritis Res Ther 2019; 21: 118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Koo MS, Manca C, Yang G, et al. Phosphodiesterase 4 inhibition reduces innate immunity and improves isoniazid clearance of mycobacterium tuberculosis in the lungs of infected mice. PLoS One 2011; 6: e17091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Subbian S, Koo MS, Tsenova L, et al. Pharmacologic inhibition of host phosphodiesterase-4 improves isoniazid-mediated clearance of mycobacterium tuberculosis. Front Immunol 2016; 7: 238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Cutolo M, Myerson GE, Fleischmann RM, et al. A phase III, randomized, controlled trial of apremilast in patients with psoriatic arthritis: results of the PALACE 2 trial. J Rheumatol 2016; 43: 1724–1734. [DOI] [PubMed] [Google Scholar]
  • 36. Edwards CJ, Blanco FJ, Crowley J, et al. Apremilast, an oral phosphodiesterase 4 inhibitor, in patients with psoriatic arthritis and current skin involvement: a phase III, randomised, controlled trial (PALACE 3). Ann Rheum Dis 2016; 75: 1065–1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Kavanaugh A, Mease PJ, Gomez-Reino JJ, et al. Treatment of psoriatic arthritis in a phase 3 randomised, placebo-controlled trial with apremilast, an oral phosphodiesterase 4 inhibitor. Ann Rheum Dis 2014; 73: 1020–1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Wells AF, Edwards CJ, Kivitz AJ, et al. Apremilast monotherapy in DMARD-naive psoriatic arthritis patients: results of the randomized, placebo-controlled PALACE 4 trial. Rheumatology (Oxford) 2018; 57: 1253–1263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Crowley J, Thaci D, Joly P, et al. Long-term safety and tolerability of apremilast in patients with psoriasis: pooled safety analysis for >/=156 weeks from 2 phase 3, randomized, controlled trials (ESTEEM 1 and 2). J Am Acad Dermatol 2017; 77: 310–317e1. [DOI] [PubMed] [Google Scholar]
  • 40. Abignano G, Fadl N, Merashli M, et al. Apremilast for the treatment of active psoriatic arthritis: a single-centre real-life experience. Rheumatology (Oxford) 2018; 57: 578–580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Favalli EG, Conti F, Selmi C, et al. Retrospective evaluation of patient profiling and effectiveness of apremilast in an Italian multicentric cohort of psoriatic arthritis patients. Clin Exp Rheumatol. Epub ahead of print 10 May 2019. [PubMed] [Google Scholar]
  • 42. Fragoulis GE, McInnes IB, Siebert S. JAK-inhibitors. New players in the field of immune-mediated diseases, beyond rheumatoid arthritis. Rheumatology (Oxford) 2019; 58(Suppl. 1): i43–i54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Boisson-Dupuis S, Ramirez-Alejo N, Li Z, et al. Tuberculosis and impaired IL-23-dependent IFN-gamma immunity in humans homozygous for a common TYK2 missense variant. Sci Immunol 2018; 3: eaau8714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Schwartz DM, Bonelli M, Gadina M, et al. Type I/II cytokines, JAKs, and new strategies for treating autoimmune diseases. Nat Rev Rheumatol 2016; 12: 25–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Casanova JL, Abel L. The human model: a genetic dissection of immunity to infection in natural conditions. Nat Rev Immunol 2004; 4: 55–66. [DOI] [PubMed] [Google Scholar]
  • 46. Abel L, El-Baghdadi J, Bousfiha AA, et al. Human genetics of tuberculosis: a long and winding road. Philos Trans R Soc Lond B Biol Sci 2014; 369: 20130428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Winthrop KL, Park SH, Gul A, et al. Tuberculosis and other opportunistic infections in tofacitinib-treated patients with rheumatoid arthritis. Ann Rheum Dis 2016; 75: 1133–1138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Dixon WG, Hyrich KL, Watson KD, et al. Drug-specific risk of tuberculosis in patients with rheumatoid arthritis treated with anti-TNF therapy: results from the British society for rheumatology biologics register (BSRBR). Ann Rheum Dis 2010; 69: 522–528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Cohen SB, Tanaka Y, Mariette X, et al. Long-term safety of tofacitinib for the treatment of rheumatoid arthritis up to 8.5 years: integrated analysis of data from the global clinical trials. Ann Rheum Dis 2017; 76: 1253–1262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Gladman D, Rigby W, Azevedo VF, et al. Tofacitinib for psoriatic arthritis in patients with an inadequate response to TNF inhibitors. N Engl J Med 2017; 377: 1525–1536. [DOI] [PubMed] [Google Scholar]
  • 51. Mease P, Hall S, FitzGerald O, et al. Tofacitinib or adalimumab versus placebo for psoriatic arthritis. N Engl J Med 2017; 377: 1537–1550. [DOI] [PubMed] [Google Scholar]
  • 52. Motoya S, Watanabe M, Kim HJ, et al. Corrigendum: tofacitinib induction and maintenance therapy in East Asian patients with active ulcerative colitis: subgroup analyses from three phase 3 multinational studies. Intest Res 2018; 16: 499–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Sandborn WJ, Ghosh S, Panes J, et al. Tofacitinib, an oral Janus kinase inhibitor, in active ulcerative colitis. N Engl J Med 2012; 367: 616–624. [DOI] [PubMed] [Google Scholar]
  • 54. Sandborn WJ, Panes J, D’Haens GR, et al. Safety of tofacitinib for treatment of ulcerative colitis, based on 4.4 years of data from global clinical trials. Clin Gastroenterol Hepatol 2019; 17: 1541–1550. [DOI] [PubMed] [Google Scholar]
  • 55. Sandborn WJ, Su C, Sands BE, et al. Tofacitinib as induction and maintenance therapy for ulcerative colitis. N Engl J Med 2017; 376: 1723–1736. [DOI] [PubMed] [Google Scholar]
  • 56. Smolen JS, Genovese MC, Takeuchi T, et al. Safety profile of baricitinib in patients with active rheumatoid arthritis with over 2 years median time in treatment. J Rheumatol 2019; 46: 7–18. [DOI] [PubMed] [Google Scholar]
  • 57. Chen YC, Yoo DH, Lee CK, et al. Safety of baricitinib in East Asian patients with moderate-to-severe active rheumatoid arthritis: an integrated analysis from clinical trials. Int J Rheum Dis 2020; 23: 65–73. [DOI] [PubMed] [Google Scholar]
  • 58. Harigai M, Takeuchi T, Smolen JS, et al. Safety profile of baricitinib in Japanese patients with active rheumatoid arthritis with over 1.6 years median time in treatment: an integrated analysis of phases 2 and 3 trials. Mod Rheumatol. Epub ahead of print 21 March 2019. DOI: 10.1080/14397595.2019.1583711. [DOI] [PubMed] [Google Scholar]
  • 59. Keystone EC, Genovese MC, Schlichting DE, et al. Safety and efficacy of baricitinib through 128 weeks in an open-label, longterm extension study in patients with rheumatoid arthritis. J Rheumatol 2018; 45: 14–21. [DOI] [PubMed] [Google Scholar]
  • 60. Burmester GR, Kremer JM, Van den Bosch F, et al. Safety and efficacy of upadacitinib in patients with rheumatoid arthritis and inadequate response to conventional synthetic disease-modifying anti-rheumatic drugs (SELECT-NEXT): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet 2018; 391: 2503–2512. [DOI] [PubMed] [Google Scholar]
  • 61. Fleischmann R, Pangan AL, Song IH, et al. Upadacitinib versus placebo or adalimumab in patients with rheumatoid arthritis and an inadequate response to methotrexate: results of a phase 3, double-blind, randomized controlled trial. Arthritis Rheumatol. Epub ahead of print 28 August 2019. DOI: 10.1002/art.41032. [DOI] [PubMed] [Google Scholar]
  • 62. Genovese MC, Fleischmann R, Combe B, et al. Safety and efficacy of upadacitinib in patients with active rheumatoid arthritis refractory to biologic disease-modifying anti-rheumatic drugs (SELECT-BEYOND): a double-blind, randomised controlled phase 3 trial. Lancet 2018; 391: 2513–2524. [DOI] [PubMed] [Google Scholar]
  • 63. Smolen JS, Pangan AL, Emery P, et al. Upadacitinib as monotherapy in patients with active rheumatoid arthritis and inadequate response to methotrexate (SELECT-MONOTHERAPY): a randomised, placebo-controlled, double-blind phase 3 study. Lancet 2019; 393: 2303–2311. [DOI] [PubMed] [Google Scholar]
  • 64. Genovese MC, Greenwald M, Codding C, et al. Peficitinib, a JAK inhibitor, in combination with limited conventional synthetic disease-modifying antirheumatic drugs in the treatment of moderate-to-severe rheumatoid arthritis. Arthritis Rheumatol 2017; 69: 932–942. [DOI] [PubMed] [Google Scholar]
  • 65. Genovese MC, Kalunian K, Gottenberg JE, et al. Effect of filgotinib vs placebo on clinical response in patients with moderate to severe rheumatoid arthritis refractory to disease-modifying antirheumatic drug therapy: the FINCH 2 randomized clinical trial. JAMA 2019; 322: 315–325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Kavanaugh A, Kremer J, Ponce L, et al. Filgotinib (GLPG0634/GS-6034), an oral selective JAK1 inhibitor, is effective as monotherapy in patients with active rheumatoid arthritis: results from a randomised, dose-finding study (DARWIN 2). Ann Rheum Dis 2017; 76: 1009–1019. [DOI] [PubMed] [Google Scholar]
  • 67. Kivitz AJ, Gutierrez-Urena SR, Poiley J, et al. Peficitinib, a JAK inhibitor, in the treatment of moderate-to-severe rheumatoid arthritis in patients with an inadequate response to methotrexate. Arthritis Rheumatol 2017; 69: 709–719. [DOI] [PubMed] [Google Scholar]
  • 68. Westhovens R, Taylor PC, Alten R, et al. Filgotinib (GLPG0634/GS-6034), an oral JAK1 selective inhibitor, is effective in combination with methotrexate (MTX) in patients with active rheumatoid arthritis and insufficient response to MTX: results from a randomised, dose-finding study (DARWIN 1). Ann Rheum Dis 2017; 76: 998–1008. [DOI] [PubMed] [Google Scholar]
  • 69. Mata-Espinosa DA, Francisco-Cruz A, Marquina-Castillo B, et al. Immunotherapeutic effects of recombinant adenovirus encoding interleukin 12 in experimental pulmonary tuberculosis. Scand J Immunol 2019; 89: e12743. [DOI] [PubMed] [Google Scholar]
  • 70. Chackerian AA, Chen SJ, Brodie SJ, et al. Neutralization or absence of the interleukin-23 pathway does not compromise immunity to mycobacterial infection. Infect Immun 2006; 74: 6092–6099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. McInnes IB, Kavanaugh A, Gottlieb AB, et al. Efficacy and safety of ustekinumab in patients with active psoriatic arthritis: 1 year results of the phase 3, multicentre, double-blind, placebo-controlled PSUMMIT 1 trial. Lancet 2013; 382: 780–789. [DOI] [PubMed] [Google Scholar]
  • 72. Ritchlin C, Rahman P, Kavanaugh A, et al. Efficacy and safety of the anti-IL-12/23 p40 monoclonal antibody, ustekinumab, in patients with active psoriatic arthritis despite conventional non-biological and biological anti-tumour necrosis factor therapy: 6-month and 1-year results of the phase 3, multicentre, double-blind, placebo-controlled, randomised PSUMMIT 2 trial. Ann Rheum Dis 2014; 73: 990–999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Kavanaugh A, Puig L, Gottlieb AB, et al. Maintenance of clinical efficacy and radiographic benefit through two years of ustekinumab therapy in patients with active psoriatic arthritis: results from a randomized, placebo-controlled phase III trial. Arthritis Care Res (Hoboken) 2015; 67: 1739–1749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Chimenti MS, Ortolan A, Lorenzin M, et al. Effectiveness and safety of ustekinumab in naive or TNF-inhibitors failure psoriatic arthritis patients: a 24-month prospective multicentric study. Clin Rheumatol 2018; 37: 397–405. [DOI] [PubMed] [Google Scholar]
  • 75. Feagan BG, Sandborn WJ, Gasink C, et al. Ustekinumab as induction and maintenance therapy for Crohn’s disease. N Engl J Med 2016; 375: 1946–1960. [DOI] [PubMed] [Google Scholar]
  • 76. Ghosh S, Gensler LS, Yang Z, et al. Ustekinumab safety in psoriasis, psoriatic arthritis, and Crohn’s disease: an integrated analysis of phase II/III clinical development programs. Drug Saf 2019; 42: 751–768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Lopez-Ferrer A, Laiz A, Puig L. The safety of ustekinumab for the treatment of psoriatic arthritis. Expert Opin Drug Saf 2017; 16: 733–742. [DOI] [PubMed] [Google Scholar]
  • 78. Tsai TF, Ho V, Song M, et al. The safety of ustekinumab treatment in patients with moderate-to-severe psoriasis and latent tuberculosis infection. Br J Dermatol 2012; 167: 1145–1152. [DOI] [PubMed] [Google Scholar]
  • 79. Hsiao CY, Chiu HY, Wang TS, et al. Serial QuantiFERON-TB gold testing in patients with psoriasis treated with ustekinumab. PLoS One 2017; 12: e0184178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Cheng C, Chung-Yee Hui R, Hu S, et al. Serial QuantiFERON-TB gold in-tube testing for psoriatic patients receiving antitumor necrosis factor-alpha therapy. Dermatol Sin 2015; 33: 124–129. [Google Scholar]
  • 81. Cuomo G, D’Abrosca V, Iacono D, et al. The conversion rate of tuberculosis screening tests during biological therapies in patients with rheumatoid arthritis. Clin Rheumatol 2017; 36: 457–461. [DOI] [PubMed] [Google Scholar]
  • 82. Cerda OL, de Los Angeles Correa M, Granel A, et al. Tuberculin test conversion in patients with chronic inflammatory arthritis receiving biological therapy. Eur J Rheumatol 2019; 6: 19–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Wu CY, Chiu HY, Tsai TF. The seroconversion rate of QuantiFERON-TB gold in-tube test in psoriatic patients receiving secukinumab and ixekizumab, the anti-interleukin-17A monoclonal antibodies. PLoS One 2019; 14: e0225112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Chen YM, Chen HH, Lai KL, et al. The effects of rituximab therapy on released interferon-gamma levels in the QuantiFERON assay among RA patients with different status of mycobacterium tuberculosis infection. Rheumatology (Oxford) 2013; 52: 697–704. [DOI] [PubMed] [Google Scholar]
  • 85. Deodhar A, Gottlieb AB, Boehncke WH, et al. Efficacy and safety of guselkumab in patients with active psoriatic arthritis: a randomised, double-blind, placebo-controlled, phase 2 study. Lancet 2018; 391: 2213–2224. [DOI] [PubMed] [Google Scholar]
  • 86. Smolen JS, Agarwal SK, Ilivanova E, et al. A randomised phase II study evaluating the efficacy and safety of subcutaneously administered ustekinumab and guselkumab in patients with active rheumatoid arthritis despite treatment with methotrexate. Ann Rheum Dis 2017; 76: 831–839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Crowley JJ, Warren RB, Cather JC. Safety of selective IL-23p19 inhibitors for the treatment of psoriasis. J Eur Acad Dermatol Venereol. Epub ahead of print 27 June 2019. DOI: 10.1111/jdv.15653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Mease PJ, Kellner H, Morita A, et al. OP0307 Efficacy and safety of risankizumab, a selective il-23p19 inhibitor, in patients with active psoriatic arthritis over 24 weeks: results from a phase 2 trial. Ann Rheum Dis 2018; 77(Suppl. 2): 200–201. [Google Scholar]
  • 89. Reich K, Gooderham M, Thaci D, et al. Risankizumab compared with adalimumab in patients with moderate-to-severe plaque psoriasis (IMMvent): a randomised, double-blind, active-comparator-controlled phase 3 trial. Lancet 2019; 394: 576–586. [DOI] [PubMed] [Google Scholar]
  • 90. Mease PJ, Chohan S, Fructuoso FJG, et al. LB0002 randomised, double-blind, placebo-controlled, multiple-dose, phase 2B study to demonstrate the safety and efficacy of tildrakizumab, a high-affinity anti-interleukin-23P19 monoclonal antibody, in patients with active psoriatic arthritis. Ann Rheum Dis 2019; 78(Suppl. 2): 78–79. [Google Scholar]
  • 91. Blauvelt A, Reich K, Papp KA, et al. Safety of tildrakizumab for moderate-to-severe plaque psoriasis: pooled analysis of three randomized controlled trials. Br J Dermatol 2018; 179: 615–622. [DOI] [PubMed] [Google Scholar]
  • 92. Hu S, He W, Du X, et al. IL-17 production of neutrophils enhances antibacteria ability but promotes arthritis development during mycobacterium tuberculosis infection. EBioMedicine 2017; 23: 88–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Singh S, Maniakis-Grivas G, Singh UK, et al. Interleukin-17 regulates matrix metalloproteinase activity in human pulmonary tuberculosis. J Pathol 2018; 244: 311–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Kammuller M, Tsai TF, Griffiths CE, et al. Inhibition of IL-17A by secukinumab shows no evidence of increased mycobacterium tuberculosis infections. Clin Transl Immunology 2017; 6: e152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Deodhar A, Mease PJ, McInnes IB, et al. Long-term safety of secukinumab in patients with moderate-to-severe plaque psoriasis, psoriatic arthritis, and ankylosing spondylitis: integrated pooled clinical trial and post-marketing surveillance data. Arthritis Res Ther 2019; 21: 111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. van de Kerkhof PC, Griffiths CE, Reich K, et al. Secukinumab long-term safety experience: a pooled analysis of 10 phase II and III clinical studies in patients with moderate to severe plaque psoriasis. J Am Acad Dermatol 2016; 75: 83–98e4. [DOI] [PubMed] [Google Scholar]
  • 97. van der Heijde D, Wei JC, Dougados M, et al. Ixekizumab, an interleukin-17A antagonist in the treatment of ankylosing spondylitis or radiographic axial spondyloarthritis in patients previously untreated with biological disease-modifying anti-rheumatic drugs (COAST-V): 16 week results of a phase 3 randomised, double-blind, active-controlled and placebo-controlled trial. Lancet 2018; 392: 2441–2451. [DOI] [PubMed] [Google Scholar]
  • 98. Mease P, Roussou E, Burmester GR, et al. Safety of ixekizumab in patients with psoriatic arthritis: results from a pooled analysis of three clinical trials. Arthritis Care Res (Hoboken) 2019; 71: 367–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Romiti R, Valenzuela F, Chouela EN, et al. Prevalence and outcome of latent tuberculosis in patients receiving ixekizumab: integrated safety analysis from 11 clinical trials of patients with plaque psoriasis. Br J Dermatol 2019; 181: 202–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Wei JC, Kim TH, Kishimoto M, et al. OP0234 efficacy and safety of brodalumab, an anti-interleukin-17 receptor a monoclonal antibody, in patients with axial spondyloarthritis: a 16 week results of a phase 3, multicenter, randomized, double-blind, placebo-controlled study. Ann Rheum Dis 2019; 78: 195. [Google Scholar]
  • 101. Mease PJ, Genovese MC, Greenwald MW, et al. Brodalumab, an anti-IL17RA monoclonal antibody, in psoriatic arthritis. N Engl J Med 2014; 370: 2295–2306. [DOI] [PubMed] [Google Scholar]
  • 102. Farahnik B, Beroukhim K, Abrouk M, et al. Brodalumab for the treatment of psoriasis: a review of phase III trials. Dermatol Ther (Heidelb) 2016; 6: 111–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Saunders BM, Frank AA, Orme IM, et al. Interleukin-6 induces early gamma interferon production in the infected lung but is not required for generation of specific immunity to Mycobacterium tuberculosis infection. Infect Immun 2000; 68: 3322–3326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Appelberg R, Castro AG, Pedrosa J, et al. Role of interleukin-6 in the induction of protective T cells during mycobacterial infections in mice. Immunology 1994; 82: 361–364. [PMC free article] [PubMed] [Google Scholar]
  • 105. Ogata A, Mori M, Hashimoto S, et al. Minimal influence of tocilizumab on IFN-gamma synthesis by tuberculosis antigens. Mod Rheumatol 2010; 20: 130–133. [DOI] [PubMed] [Google Scholar]
  • 106. Cantini F, Nannini C, Niccoli L, et al. Risk of tuberculosis reactivation in patients with rheumatoid arthritis, ankylosing spondylitis, and psoriatic arthritis receiving non-anti-TNF-targeted biologics. Mediators Inflamm 2017; 2017: 8909834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Cantini F, Niccoli L, Goletti D. Tuberculosis risk in patients treated with non-anti-tumor necrosis factor-alpha (TNF-alpha) targeted biologics and recently licensed TNF-alpha inhibitors: data from clinical trials and national registries. J Rheumatol Suppl 2014; 91: 56–64. [DOI] [PubMed] [Google Scholar]
  • 108. Stone JH, Tuckwell K, Dimonaco S, et al. Trial of tocilizumab in giant-cell arteritis. N Engl J Med 2017; 377: 317–328. [DOI] [PubMed] [Google Scholar]
  • 109. Villiger PM, Adler S, Kuchen S, et al. Tocilizumab for induction and maintenance of remission in giant cell arteritis: a phase 2, randomised, double-blind, placebo-controlled trial. Lancet 2016; 387: 1921–1927. [DOI] [PubMed] [Google Scholar]
  • 110. Nisar MK, Rafiq A, Ostor AJ. Biologic therapy for inflammatory arthritis and latent tuberculosis: real world experience from a high prevalence area in the United Kingdom. Clin Rheumatol 2015; 34: 2141–2145. [DOI] [PubMed] [Google Scholar]
  • 111. Tarkiainen M, Tynjala P, Vahasalo P, et al. Occurrence of adverse events in patients with JIA receiving biologic agents: long-term follow-up in a real-life setting. Rheumatology (Oxford) 2015; 54: 1170–1176. [DOI] [PubMed] [Google Scholar]
  • 112. Shobha V, Chandrashekara S, Rao V, et al. Biologics and risk of tuberculosis in autoimmune rheumatic diseases: a real-world clinical experience from India. Int J Rheum Dis 2019; 22: 280–287. [DOI] [PubMed] [Google Scholar]
  • 113. Sakai R, Cho SK, Nanki T, et al. Head-to-head comparison of the safety of tocilizumab and tumor necrosis factor inhibitors in rheumatoid arthritis patients (RA) in clinical practice: results from the registry of Japanese RA patients on biologics for long-term safety (REAL) registry. Arthritis Res Ther 2015; 17: 74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Lim CH, Chen HH, Chen YH, et al. The risk of tuberculosis disease in rheumatoid arthritis patients on biologics and targeted therapy: a 15-year real world experience in Taiwan. PLoS One 2017; 12: e0178035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Lin CT, Huang WN, Hsieh CW, et al. Safety and effectiveness of tocilizumab in treating patients with rheumatoid arthritis - a three-year study in Taiwan. J Microbiol Immunol Infect 2019; 52: 141–150. [DOI] [PubMed] [Google Scholar]
  • 116. Yonekura CL, Oliveira RDR, Titton DC, et al. Incidence of tuberculosis among patients with rheumatoid arthritis using TNF blockers in Brazil: data from the Brazilian registry of biological therapies in rheumatic diseases (Registro Brasileiro de monitoracao de terapias biologicas - biobadaBrasil). Rev Bras Reumatol Engl Ed 2017; 57(Suppl. 2): 477–483. [DOI] [PubMed] [Google Scholar]
  • 117. Pawar A, Desai RJ, Solomon DH, et al. Risk of serious infections in tocilizumab versus other biologic drugs in patients with rheumatoid arthritis: a multidatabase cohort study. Ann Rheum Dis 2019; 78: 456–464. [DOI] [PubMed] [Google Scholar]
  • 118. Tan BE, Lim AL, Kan SL, et al. Real-world clinical experience of biological disease modifying anti-rheumatic drugs in Malaysia rheumatoid arthritis patients. Rheumatol Int 2017; 37: 1719–1725. [DOI] [PubMed] [Google Scholar]
  • 119. Koike T, Harigai M, Inokuma S, et al. Postmarketing surveillance of tocilizumab for rheumatoid arthritis in Japan: interim analysis of 3881 patients. Ann Rheum Dis 2011; 70: 2148–2151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Lee EB. A review of sarilumab for the treatment of rheumatoid arthritis. Immunotherapy 2018; 10: 57–65. [DOI] [PubMed] [Google Scholar]
  • 121. Sieper J, Braun J, Kay J, et al. Sarilumab for the treatment of ankylosing spondylitis: results of a phase II, randomised, double-blind, placebo-controlled study (ALIGN). Ann Rheum Dis 2015; 74: 1051–1057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Weinblatt ME, Mease P, Mysler E, et al. The efficacy and safety of subcutaneous clazakizumab in patients with moderate-to-severe rheumatoid arthritis and an inadequate response to methotrexate: results from a multinational, phase IIb, randomized, double-blind, placebo/active-controlled, dose-ranging study. Arthritis Rheumatol 2015; 67: 2591–2600. [DOI] [PubMed] [Google Scholar]
  • 123. Mease PJ, Gottlieb AB, Berman A, et al. The efficacy and safety of clazakizumab, an anti-interleukin-6 monoclonal antibody, in a phase IIb study of adults with active psoriatic arthritis. Arthritis Rheumatol 2016; 68: 2163–2173. [DOI] [PubMed] [Google Scholar]
  • 124. Aletaha D, Bingham CO, III, Tanaka Y, et al. Efficacy and safety of sirukumab in patients with active rheumatoid arthritis refractory to anti-TNF therapy (SIRROUND-T): a randomised, double-blind, placebo-controlled, parallel-group, multinational, phase 3 study. Lancet 2017; 389: 1206–1217. [DOI] [PubMed] [Google Scholar]
  • 125. Takeuchi T, Thorne C, Karpouzas G, et al. Sirukumab for rheumatoid arthritis: the phase III SIRROUND-D study. Ann Rheum Dis 2017; 76: 2001–2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Takeuchi T, Yamanaka H, Harigai M, et al. Sirukumab in rheumatoid arthritis refractory to sulfasalazine or methotrexate: a randomized phase 3 safety and efficacy study in Japanese patients. Arthritis Res Ther 2018; 20: 42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Taylor PC, Schiff MH, Wang Q, et al. Efficacy and safety of monotherapy with sirukumab compared with adalimumab monotherapy in biologic-naive patients with active rheumatoid arthritis (SIRROUND-H): a randomised, double-blind, parallel-group, multinational, 52-week, phase 3 study. Ann Rheum Dis 2018; 77: 658–666. [DOI] [PubMed] [Google Scholar]
  • 128. Weinblatt ME, Moreland LW, Westhovens R, et al. Safety of abatacept administered intravenously in treatment of rheumatoid arthritis: integrated analyses of up to 8 years of treatment from the abatacept clinical trial program. J Rheumatol 2013; 40: 787–797. [DOI] [PubMed] [Google Scholar]
  • 129. Takahashi N, Kojima T, Kaneko A, et al. Longterm efficacy and safety of abatacept in patients with rheumatoid arthritis treated in routine clinical practice: effect of concomitant methotrexate after 24 weeks. J Rheumatol 2015; 42: 786–793. [DOI] [PubMed] [Google Scholar]
  • 130. Salmon JH, Gottenberg JE, Ravaud P, et al. Predictive risk factors of serious infections in patients with rheumatoid arthritis treated with abatacept in common practice: results from the orencia and rheumatoid arthritis (ORA) registry. Ann Rheum Dis 2016; 75: 1108–1113. [DOI] [PubMed] [Google Scholar]
  • 131. Gottenberg JE, Ravaud P, Bardin T, et al. Risk factors for severe infections in patients with rheumatoid arthritis treated with rituximab in the autoimmunity and rituximab registry. Arthritis Rheum 2010; 62: 2625–2632. [DOI] [PubMed] [Google Scholar]
  • 132. Tony HP, Burmester G, Schulze-Koops H, et al. Safety and clinical outcomes of rituximab therapy in patients with different autoimmune diseases: experience from a national registry (GRAID). Arthritis Res Ther 2011; 13: R75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Xanthouli P, Sailer S, Fiehn C. Rituximab (RTX) as an alternative to TNF-alpha antagonists in patients with rheumatoid arthritis and high risk of severe infections:a systematic analysis of the experience in one center. Open Rheumatol J 2012; 6: 286–289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Wendler J, Burmester GR, Sorensen H, et al. Rituximab in patients with rheumatoid arthritis in routine practice (GERINIS): six-year results from a prospective, multicentre, non-interventional study in 2,484 patients. Arthritis Res Ther 2014; 16: R80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Liao TL, Lin CH, Chen YM, et al. Different risk of tuberculosis and efficacy of isoniazid prophylaxis in rheumatoid arthritis patients with biologic therapy: a nationwide retrospective cohort study in Taiwan. PLoS One 2016; 11: e0153217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Bigbee CL, Gonchoroff DG, Vratsanos G, et al. Abatacept treatment does not exacerbate chronic mycobacterium tuberculosis infection in mice. Arthritis Rheum 2007; 56: 2557–2565. [DOI] [PubMed] [Google Scholar]
  • 137. van Vollenhoven RF, Emery P, Bingham CO, III, et al. Long-term safety of rituximab in rheumatoid arthritis: 9.5-year follow-up of the global clinical trial programme with a focus on adverse events of interest in RA patients. Ann Rheum Dis 2013; 72: 1496–1502. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Therapeutic Advances in Musculoskeletal Disease are provided here courtesy of SAGE Publications

RESOURCES