Next Article in Journal
Entomopathogenic Potential of Bacillus velezensis CE 100 for the Biological Control of Termite Damage in Wooden Architectural Buildings of Korean Cultural Heritage
Next Article in Special Issue
Development of a Prediction Model for Short-Term Remission of Patients with Crohn’s Disease Treated with Anti-TNF Drugs
Previous Article in Journal
Genetics and Epigenetics in Complex Diseases
Previous Article in Special Issue
90K/Mac-2 BP Is a New Predictive Biomarker of Response to Infliximab Therapy in IBD Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Focus on Anti-Tumour Necrosis Factor (TNF)-α-Related Autoimmune Diseases

by
Loris Riccardo Lopetuso
1,2,3,
Claudia Cuomo
1,
Irene Mignini
1,
Antonio Gasbarrini
1,4 and
Alfredo Papa
1,4,*
1
Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Gemelli, IRCCS, 00168 Rome, Italy
2
Department of Medicine and Ageing Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
3
Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
4
Department of Translational Medicine and Surgery, School of Medicine, Catholic University, 00168 Rome, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(9), 8187; https://doi.org/10.3390/ijms24098187
Submission received: 15 April 2023 / Revised: 1 May 2023 / Accepted: 2 May 2023 / Published: 3 May 2023
(This article belongs to the Special Issue Molecular Research in Inflammatory Bowel Disease)

Abstract

:
Anti-tumour necrosis factor (TNF)-α agents have been increasingly used to treat patients affected by inflammatory bowel disease and dermatological and rheumatologic inflammatory disorders. However, the widening use of biologics is related to a new class of adverse events called paradoxical reactions. Its pathogenesis remains unclear, but it is suggested that cytokine remodulation in predisposed individuals can lead to the inflammatory process. Here, we dissect the clinical aspects and overall outcomes of autoimmune diseases caused by anti-TNF-α therapies.

1. Introduction

Anti-tumour necrosis factor (TNF)-α agents are increasingly being used for the treatment of patients affected by inflammatory bowel disease (IBD) such as ulcerative colitis (UC) and Crohn’s disease (CD), as well as rheumatologic and dermatological disorders such as rheumatoid arthritis (RA), juvenile idiopathic arthritis (JIA), ankylosing spondylitis (AS), psoriatic arthritis (PsA), and psoriasis. Notably, the majority of these drugs can neutralise both forms of TNF-α, either by binding the trans-membrane form (m-TNF-α) or by blocking the soluble form (s-TNF-α) [1]. Only infliximab (IFX) and adalimumab (ADA) also induce apoptosis in T cells and monocytes [1,2]. Conversely, etanercept (ETA) is a recombinant, covalently bound dimer of the soluble p75 TNF receptor fused to the Fc portion IgG1, which binds to the soluble TNF-α form, but not the membrane-bound TNF-α [3]. Among the TNF-α blockers, only ETA is not approved for treating IBD patients, both CD and UC. In contrast, the last authorised agent, golimumab (GOL), is given only for UC but not for CD, other than for other clinical conditions such as RA, PsA, AS, and polyarticular JIA. On the other hand, the fifth anti-TNF-α agent, certolizumab pegol (CER), is only approved for CD by the United States Food and Drug Administration (FDA) but not by the European Medicines Agency (EMA), though it is used for PsA, RA, SpA, and psoriasis. It is worth noting that ETA, GOL, CER, and ADA are given subcutaneously, while IFX is administered intravenously. However, it is worth mentioning that subcutaneous IFX has recently become available, but there is no data yet on possible paradoxical autoimmune manifestations involving it.
With the widespread use of anti-TNF-α agents, concerns about any unexpected effects have been raised. Indeed, over the past 20 years, the widening use of biologics in several autoimmune diseases has been related to a new class of adverse events called paradoxical reactions. These immune-mediated processes appear paradoxically during treatment and can virtually affect several organs, including the skin, liver, lungs, kidneys, peripheral and central nervous system, vascular system, and bowels (Table 1). Despite the significant role of these drugs in suppressing TNF-α, a crucial cytokine in the development of the inflammatory process, a growing number of reports of paradoxical autoimmune processes associated with anti-TNF-α agents have emerged. This review aims to dissect the clinical aspects and overall outcomes of autoimmune diseases caused by biological therapies.

2. Dermatological Manifestations

Anti-TNF-α therapy can cause a wide range of dermatological conditions, including local skin irritation or reaction, increased skin infection rates, psoriasis, eczema, anti-TNF-induced cutaneous lupus erythematosus (ATIL), and alopecia areata (AA). Other reactions rarely occur, such as granuloma annulare, lichen planus, vitiligo, and cutaneous vasculitis. In patients with IBD, some of the above complications, such as erythema nodosum and pyoderma gangrenosum, could be extraintestinal manifestations related or not to clinical exacerbations. Thus, temporal associations with biological therapy could differentiate between disease- and drug-related complications. These reactions are managed by stopping anti-TNF-α treatment, switching to a different TNF-α inhibitor or ustekinumab, switching to another systemic immunosuppressant for treating the underlying dermatologic condition, or continuing anti-TNF-α therapy with the addition of topical or systemic skin-directed treatments, depending on the severity and extension of the cutaneous lesions.

2.1. Psoriasis

Psoriasis is a chronic autoimmune disease characterised by raised red, itchy, scaly patches, most commonly on the knees, elbows, trunk, and scalp. TNF-α antagonist-induced psoriasis, which can emerge de novo or exacerbate a pre-existing form, is the most common dermatological adverse reaction linked to anti-TNF therapy [4,5]. In most cases (80%), these adverse events represent a new disease, while others exacerbate a pre-existing illness. In a recent case series involving 150 patients with psoriasis who received treatment with TNF-α inhibitors [6], 10 patients developed paradoxical psoriasis (PP) without stopping anti-TNF-α therapy. Nonetheless, 80% of them achieved remission again after the aggravation of symptoms without any other serious adverse events. Several studies have been conducted to identify the main characteristics and pathogenesis of these specific lesions, concluding that the pathogenesis of PP could be different from that of classical psoriasis. In fact, it has been shown that classical psoriasis is a T-cell-mediated autoimmune disease driven by TNF.
In contrast, PP is caused by the absence of TNF-α and represents type I interferon (IFN)-driven innate inflammation [7]. Stoffel sought to characterise anti-TNF-induced inflammatory skin lesions on a histopathologic, cellular, and molecular level compared to psoriasis, eczema (atopic dermatitis), and healthy control skin [8]. Histopathologic evaluation, gene expression, and computer-assisted immunohistologic studies have been performed on 19 skin biopsies from IBD (n = 17) and rheumatoid arthritis (n = 2) patients with new-onset inflammatory skin lesions during anti-TNF-α therapy [8], evidencing the new onset of psoriasiform, eczematous, and scalp anti-TNF-induced skin lesions. While the histopathologic aspects were similar, these reactions were considered distinct immunological entities with Th1/IFN-γ/IFN-α characteristics; all the anti-TNF-induced lesions showed a more robust IFN-γ activation than in psoriasis or eczema [8]. This signature was consistent with an increased concentration of Th1 but not Tc1 or natural killer (NK) cells. Another study investigated the immunological and genetic profiles of three hidradenitis suppurativa (HS) patients without psoriasis who developed paradoxical psoriasiform reactions following anti-TNF-α therapy with ADA. These skin manifestations showed immunological features common to the early phases of psoriasis [9]. Among them, LT-α and LT-β, as well as IFN-κ and IFN-λ1, were identified as new innate mediators potentially involved in the induction process, mainly characterised by innate cellular and molecular players [9].
IFX and ADA are the most frequently associated with psoriasis among anti-TNF drugs. Collamer et al. reviewed 207 cases of new-onset psoriasis, of which 43% of patients had RA, 26% seronegative SpA, and 20% IBD [10]. The most frequently implicated drug was IFX (50% of cases), followed by ADA and ETA, while reactions to Certolizumab or Golimumab were infrequent [10]. In this scenario, in IBD, the onset of PP has been associated with the female sex, comorbidities, and the use of ADA [11]. In addition, a high incidence of anti-TNF-associated psoriasis in patients treated with IFX or ADA was confirmed in further analyses, which also documented an association with female gender, foregut disease location, fistulising, and stricturing disease behaviour [12,13].
Besides the adult setting, little is known about paediatric IBD patients. PP appears to be the most frequent side effect in paediatric CD treated with IFX. Perianal lesions and young age at anti-TNF initiation represent the most significant risk factors [14].

2.2. Anti-TNF-Induced Lupus (ATIL)

Following the introduction of anti-TNF, ATIL has become a new and increasingly recognised clinical entity [15]. All anti-TNF-α agents have been associated with an increased risk of autoantibodies. Up to 49% of patients on these medications develop raised antinuclear antibodies (ANAs) or anti-double strain (ds) DNA. However, despite the high antibody prevalence, clinical manifestations of ATIL remain rare [16].
Overall, the first case of drug-induced lupus (DILE) was reported in 1945 [17] as a complication of sulfasalazine therapy, described as a milder disease form than classical systemic lupus erythematosus (SLE), which resolved spontaneously after stopping treatment. DILE is an autoimmune disorder that can present with joint pain, skin involvement, fatigue, and serositis. Conversely, ATIL appears distinct from DILE, with a phenotype similar to idiopathic SLE. The incidence/prevalence of dsDNA antibodies and hypocomplementemia is greater in ATIL, while anti-histone antibodies, the serological hallmark of classical DILE, are less commonly found. The most common symptoms of ATIL are arthralgias and arthritis, haematological abnormalities, and a skin involvement similar to SLE [18]. Cerebral and renal involvement has been reported more frequently in ATIL than in classical DILE. In this scenario, the central nervous system (CNS) is not often involved, and neuropsychiatric symptoms such as depression and suicidal ideations are not commonly seen in ATIL patients [19].
A large Australian cohort study including 454 patients treated with anti-TNF-α therapy (300 with IFX and 154 with ADA) reported an incidence of ATIL of 5.7% for IFX and 0.6% for ADA [20]. These rates appear much higher than those in post-marketing studies. The diagnostic criteria for ATIL diagnosis are a temporal relationship between symptoms and anti-TNF-α therapy administration and resolution of symptoms following cessation of the offending medication; at least one serologic American College of Rheumatology criterion of SLE; at least one non-serological criterion such as arthritis, serositis, or a rash. Another dedicated study assessed the clinical characteristics of three IBD patients treated with IFX [21]. The first patient had a classical DILE with thrombocytopenia that resolved after IFX discontinuation, the second case experienced symmetric polyarthritis of 14 joints in RA-like distribution accompanied by lymphopenia, and the third one had severe serositis with ascites, pleural and pericardial effusions, and pancytopenia [21]. In this patient, ATIL coexisted with anti-TNF-induced hepatitis [21]. The occurrence of both lupus-like syndrome and hepatitis following anti-TNF-α therapy in the same patient is infrequent. According to these reports, the clinical presentations of ATIL can vary from mild cases to severe adverse events. However, specific diagnostic criteria for ATIL have not been recognised yet. Although the primary evidence of lupus development comes from studies involving IBD patients, this feature is not exclusive since it can also occur in other chronic inflammatory conditions [22]. Of note, while stopping anti-TNF-α therapy has. been shown to be beneficial, reintroducing another anti-TNF-α does not cause significant consequences [23]. Only a few severe cases characterised by systemic involvement have required conventional therapy for SLE to achieve complete remission.

2.3. Other Skin Manifestations

Alopecia areata (AA) can be described as a non-scarring inflammatory pathology with a sudden onset and rapid hair loss in some regions of the scalp and body. It can be divided into alopecia focalis (monolocularis or multilocularis), which can occur in one or multiple areas of the scalp; alopecia totalis, which happens all over the scalp; alopecia universalis, which is a complete loss of scalp and body hair. It is linked to a multifactorial process involving an immuno-mediated response and the overexpression of proinflammatory cytokines such as TNF-α, interleukin-1-α, and IFN-γ. According to this evidence, a role for anti-TNF in the pathogenesis of AA seems unlikely. Nevertheless, few reports have shown this connection [24], such as the case of a 50-year-old man with psoriatic arthritis who exhibited alopecia universalis with concomitant onychodystrophy three months after ADA initiation [25]. In this setting, anti-TNF therapy could have caused cytokine dysregulation that could trigger these manifestations.
Among dermatological adverse effects, reports have associated TNF-α inhibitors with the onset of lichenoid eruption (LE). Lichen planus is a recurrent, itchy inflammatory rash with single, small, polygonal papules that can merge to form rough, scaly plaques, often accompanied by injury to the oral cavity or genitals. Diagnosis is usually clinical and can be supported by a skin biopsy. Treatment generally requires topical or intralesional corticosteroids. A case report described three IBD patients who developed oral lichen planus after starting anti-TNF therapy [26]. Another paper documented five patients with LE following anti-TNF-α treatment for ankylosing spondylitis [27]. In these cases, the development of an inflammatory reaction is somewhat surprising since TNF-α is involved in the pathogenesis of lichen planus, and TNF-α inhibitors are efficacious when used to treat refractory cases. The potential cause could be linked to the anti-TNF-mediated remodulation of cytokine release with subsequent increasing levels of type I IFN and the activation of T and dendritic cells [27].
Vitiligo is a long-term skin condition characterised by patches of the skin losing its pigment. Hypotheses for its pathogenesis include immune-mediated destruction of epidermal melanocytes [28]. Vitiligo treatment involves general, non-targeted immunosuppressants that provide only modest efficacy. TNF-α has been shown to contribute to depigmentation. Indeed, TNF-α levels in vitiligo lesions are higher than those in non-lesional skin and are closely related to disease activity [29,30]. Following this rationale, anti-TNF treatments have been described for active vitiligo.
Conversely, new-onset cases following biological treatment have emerged [31]. In this setting, the risk of developing vitiligo and AA in patients with AS, CD, or UC treated with anti-TNF therapy compared to those without anti-TNF agents has been evaluated using the Korea National Health Insurance Claims databases [32]. A significantly increased risk of vitiligo was observed in the anti-TNF-α group compared to the unexposed group. In subgroup analyses, younger patients and those treated with ETA showed the highest risk. No statistically significant difference was observed in the development of AA between the two groups. Accordingly, TNF inhibition may shift cytokine patterns with the subsequent recruitment of autoreactive T cells to the epidermis and the destruction of melanocytes.
In addition, several cases of anti-TNF-mediated dermatomyositis/polymyositis have been described [33]. Here, the pathophysiological mechanism is unknown, but increasing interferon-gamma levels following therapy could represent a crucial risk factor [34].

3. Liver Diseases

The primary concerns about anti-TNF-α therapy-related liver injury refer to the increased susceptibility towards chronic infections, especially towards the reactivation of the hepatitis B virus (HBV) [35]. Autoimmune hepatic toxicity is less common, but several cases have been reported during post-marketing surveillance. Although not frequent, autoimmune disorders are nonetheless significant [35,36]. The drug most often involved is IFX and, to a lesser extent, ADA and ETA, with a solid female predominance [36,37]. The predominant form of liver injury mediation is characterised by a hepatocellular profile with both serological and histological features of autoimmunity. In most reported cases, biopsy shows chronic lymphoplasmocytic infiltrate and interface hepatitis with altered liver function tests associated with positive autoantibodies, such as ANA, anti-smooth muscle antibodies (ASMAs) or anti-dsDNA, or increased immunoglobulins [38]. These findings support the autoimmune origin of anti-TNF-related hepatitis, which resembles the autoimmune hepatitis type I or hepatitis in the context of a lupus-like syndrome. Moreover, anti-TNF-induced liver diseases include hepatocellular injury without autoimmunity and cholangiopathy [39,40].
Clinical manifestations range from asymptomatic abnormalities in liver function tests, self-limiting after drug discontinuation, to more severe damage, requiring corticosteroid therapy. Tobon et al. described two cases of severe liver disease: a 39-year-old woman who was treated with IFX for eight months and developed cholestatic liver disease and hepatic failure with hepatic encephalopathy and ascites and finally underwent liver transplantation, and a 54-year-old woman who was diagnosed with acute hepatitis with positive ANAs after 17 months of IFX therapy and was treated with prednisolone and azathioprine [41]. Cases of fulminant hepatitis have also been reported, though rarely, leading to transplantation or death. In clinical practice, differentiating between autoimmune hepatitis and drug-induced hepatitis is challenging, with often indistinguishable symptoms, serology, and histology. Usually, patients with anti-TNF-induced liver injury do not relapse after the resolution, with or without immunosuppressive therapy, and ANAs disappear after steroid therapy [42,43]. Here, the pathogenesis is still unclear. However, it has been hypothesised to be more complex than just a class effect since some patients with IFX-mediated autoimmune hepatitis well tolerated other TNF-α inhibitors [35,36,44,45]. It has been suggested that the absence of cross-toxicity between anti-TNF-α drugs and differences in the clinical response may be due to their different molecular structure. This hypothesis could also explain why IFX, the only chimeric mouse–human antibody, is the anti-TNF-α most prone to causing autoimmune liver injury [46]. Thus, a pathogenic role could be played by specific antibodies against IFX. Moreover, IFX could impair the physiological suppression of auto-reactive B cells, leading to an increased lymphocyte presence. It may provoke cellular apoptosis and elicit self-antigens release, causing the loss of tolerance towards the patient’s hepatocytes [35,36,47,48].

4. Interstitial Lung Diseases

Anti-TNF-related lung autoimmune disorders are mainly represented by interstitial lung diseases (ILDs) and pulmonary fibrosis, the most severe manifestation. However, the association between these drugs and developing or exacerbating ILDs is still controversial [49]. Experimental evidence shows that TNF-α antagonists may improve or paradoxically induce ILDs [49,50]. ILDs include heterogeneous lung disorders with essential and potentially severe manifestations of chronic inflammation, ranging from asymptomatic to potentially life-threatening. Even if an ideal treatment is not well established, immunosuppressive drugs and TNF-α blockers are administrated to reduce inflammation and stabilise the progression of ILDs [51].
In contrast, case reports have described ILDs and pulmonary fibrosis as paradoxical side effects of anti-TNF-α agents. These drugs may both induce new lung injury and worsen pre-existing ILDs. Most available data refer to patients affected by rheumatoid arthritis [52,53]. The drugs most frequently involved are IFX and ETA, followed by ADA. Interstitial pneumonia appears to be the most common pulmonary autoimmune complication of these drugs [54,55,56]. Interestingly, Komiya et al. reported the case of a man with a history of RA and lung involvement. His pulmonary lesions and arthritic symptoms first improved with ADA, but then he developed interstitial pneumonia, showing the simultaneous conflicting actions of anti-TNF agents [57].
Similarly, even if less frequently, developing or exacerbating ILDs under biological therapy has also been reported in diseases other than rheumatoid arthritis, such as psoriatic arthritis/psoriasis, ankylosing spondylitis, systemic sclerosis, and IBD [55,58,59]. The case of a 53-year-old man with UC was described in this setting. He reported a fatal acute interstitial pneumonitis after completing an accelerated induction course of IFX. Four similar cases of IFX-induced interstitial lung disease in UC patients were also evidenced, but they had a successful outcome after IFX discontinuation [60]. However, a large cohort study conducted on 8471 patients with RA, ankylosing spondylitis, psoriatic arthritis, psoriasis, and IBD showed that anti-TNF-α therapy does not increase the occurrence of ILDs among patients with autoimmune diseases when compared to non-biologic treatments [61].
Conversely, it has been suggested that anti-TNF-α might exacerbate pre-existing ILDs compared to non-anti-TNF-α biological therapy [62]. In a cohort of 40 patients, most cases showed that anti-TNF-α was harmful to patients with ILD, with a 35% mortality rate. Age, use of antibiotics, and association with azathioprine were considered risk factors. In addition, adverse ILD events tended to appear in female patients with a long history of RA. The most common clinical manifestations in ILD adverse events were shortness of breath and sudden dyspnoea, followed by non-productive cough [63]. Radiologically, the non-specific interstitial pneumonia pattern was the most common imaging type [63]. Translational studies have demonstrated that despite a pathogenic role of TNF-α in favouring chronic lymphocytic alveolitis [64] and fibroblast activation [65], and vice versa, a lack of TNF-α seems to prevent bleomycin-induced lung fibrosis [66]. Further studies are needed to clarify the uncertain role of TNF-α inhibitors in lung disorders and help treatment decisions. In particular, more caution should be taken when prescribing anti-TNF to RA patients, especially older patients with a more extended RA history or RA-associated pulmonary disease.

5. Renal Disorders

Compared to other anti-TNF-mediated adverse events, autoimmune renal disorders are uncommon. They are most often described in patients affected by rheumatic diseases, especially rheumatoid arthritis, ankylosing spondylitis, and psoriatic arthritis, but also in other conditions, such as psoriasis and IBD. ETA is the drug most frequently implicated, followed by ADA and IFX [67]. Renal alterations can appear as isolated manifestations or as part of more complex disorders such as systemic vasculitis or SLE. Different types of vasculitis with renal involvement have been described, notably ANCA-associated vasculitis, both p-ANCA and c-ANCA, which are often characterised by a histopathological pattern of necrotising crescentic glomerulonephritis [68,69], and Henoch–Schönlein purpura, which are often related to mesangial glomerulonephritis [70]. Biopsies performed in SLE-associated glomerulonephritis have shown class IV or, less frequently, class III lupus glomerulonephritis. These patients had increased levels of antinuclear antibodies (ANAs), and some also had increased anti-dsDNA antibodies [71]. Isolated renal disorders show various manifestations, including membranous, mesangial, necrotising crescentic, minimal changes in glomerulonephritis, and focal segmental glomerulosclerosis [72].
Acute non-granulomatous tubulointerstitial nephritis has also been reported in an HLA-B27-positive patient with axial spondylarthritis and CD treated with ADA [73]. Sporadic cases of renal sarcoidosis have also been reported [74,75].

6. Systemic and Cutaneous Vasculitis

An increasing number of therapeutic agents have been associated with vasculitic syndromes. Among these, TNF-α inhibitors play a central role. Cutaneous manifestations with palpable purpura and leukocytoclastic vasculitis (LCV) are usually the most common upon histological examination, while systemic vasculitis mainly affects the peripheral nervous system and the kidneys [76,77,78]. Other sites of vasculitic involvement can include the lungs, central nervous system, gallbladder, and coronary arteries. LCV is an uncommon small-vessel vasculitis known as hypersensitivity vasculitis and hypersensitivity angiitis. It is an inflammatory process that can affect the small blood vessels of any organ. When acute, cutaneous leukocytoclastic vasculitis is the most frequent feature. Potential LCV causes include autoimmune diseases (i.e., RA, SLE, Sjogren, and Henoch–Schönlein purpura), infection, and drug-mediated immune reactions, especially TNF-α inhibitors. Some authors have suggested that host antibodies developed against anti-TNF-α agents may cause an immune-complex-mediated hypersensitivity reaction [79]. In most cases, drug discontinuation and sometimes corticosteroid treatment lead to the complete resolution of the cutaneous manifestations. In this setting, individual genetic susceptibility could explain the different disease localisations between cutaneous and systemic vasculitis.

7. Neuropathy

Demyelination has been described as a consequence of anti-TNF-α therapy [80]. A review of the FDA Adverse Event Reporting System showed 772 reports of neurological complications, especially in patients with RA (50.9%) and IBD (18%). ETA, followed by IFX, resulted in the most involved drugs [81]. Peripheral neuropathy was the most frequent manifestation, followed by CNS and spinal cord demyelination [81,82]. However, CNS demyelination after anti-TNF-α treatment is not necessarily associated with the duration of therapy and drug discontinuation does not always coincide with the resolution of symptoms.

8. Sarcoidosis

Sarcoidosis is a systemic inflammatory disorder characterised by non-caseating granuloma, which commonly affects the lungs, lymph nodes, and salivary glands, but rarely other organs (i.e., liver, spleen, eyes, heart, and skin). In recent years, TNF-α inhibitors have been used for its treatment. However, there have been isolated cases of sarcoidosis occurrence during anti-TNF therapy. Daïen et al. estimated a frequency of 1:2800 [83]. In most cases, sarcoidosis occurs after treatment with ETA [83], but it has also been described with ADA and IFX [84,85,86]. The underlying disease is often RA, followed by AS and PsA. Symptom improvement is registered in most patients after drug exclusion, with only a few cases requiring steroid therapy [84,85,86,87,88].
Interestingly, Isshiki et al. described the case of an RA patient who developed sarcoidosis six months after the introduction of ETA and improved without therapeutic intervention after its cessation [89]. In this case, Propionibacterium acnes was detected in the granulomas, suggesting that P. acnes infection may show aberrant responses to drugs such as TNF-α inhibitors and immune checkpoint inhibitors, consequently leading to drug-induced sarcoidosis [89]. Further potential hypotheses on the pathogenesis of anti-TNF-mediated sarcoidosis have been proposed. It has been shown that the mechanisms of action of ETA and IFX can produce different TNF-α concentrations in the tissue, and higher levels of biologically active TNF-α from ETA therapy may foster the formation of granulomas [79]. This phenomenon may be due to the molecular structure of ETA. While monoclonal antibodies (i.e., IFX and ADA) can neutralise both soluble and membrane forms of TNF-α, ETA neutralises just the soluble form. Only “partial” inhibition can lead to a redistribution of TNF-α in areas of elevated concentration, such as the lungs and lymph nodes [90,91].
Furthermore, several studies have suggested that monoclonal antibodies, in contrast to ETA, inhibit the expression of the IL-17 and IFN-γ cytokines intensely involved in granuloma formation. In contrast, some reports have shown sarcoid-like reactions in patients treated with IFX and ADA [92,93]. In these cases, cytokine imbalance due to long-term TNF-α suppression could lead to those paradoxical reactions. Moreover, as shown for paradoxical skin reactions, anti-TNF-α can increase INF production, which is crucial for granuloma formation [94].

9. New-Onset IBD

Both paradoxical worsening of IBD and new-onset cases induced by anti-TNF-α agents have been reported in patients with different inflammatory diseases, especially AS, JIA, and psoriasis. CD has been observed more frequently than UC, with most cases attributed to ETA [95,96]. In contrast with the monoclonal antibodies, ETA is ineffective in treating IBD because of its inability to induce T cell apoptosis in the intestinal mucosa [97]. Some authors have suggested that paradoxical IBD could represent a subclinical condition unmasked by TNF-α inhibitors, particularly by ETA and less frequently by IFX [96,98,99,100]. The reason behind this different risk rate is again linked to their divergent mechanisms of action. IFX binds to both soluble and membrane TNF-α, while ETA binds to soluble TNF-α and lymphotoxin-α, causing a partial TNF-α neutralisation. IFX but not ETA can induce circulating and lamina propria T lymphocyte apoptosis by binding directly to transmembrane TNF-α and caspase-3 activation. Conversely, ETA can increase T lymphocyte levels in peripheral blood and the number of proinflammatory cytokines, such as IFN-γ, which can trigger intestinal inflammation. Despite the well-known concept that extraintestinal inflammatory diseases (i.e., AS, JIA, psoriasis, and PsA) can represent independent risk factors for the development of IBD, the fact that, after the interruption of ETA and the switch to a different anti-TNF-α or the swap to another biological agent, patients usually obtain an immediate improvement of gastrointestinal inflammation suggests the theory of a paradoxical effect [101,102].

10. Conclusions

Anti-TNF-α agents has significantly improved the outcomes of IBD and dermatological and rheumatological conditions. Paradoxical events due to these therapies are not commonly reported in the literature, especially compared to other side effects. In recent years, a growing number of cases and a more extensive range of clinical manifestations have been reported because of their increased use. The pathogenesis of the paradoxical reactions is not completely clarified yet. However, several mechanisms involved have been hypothesised and are summarised in Table 2. They include cytokine remodulation, which leads to the overexpression of some inflammatory molecules (i.e., IFN and IL-17/IL-23) able to trigger an inflammatory process, promoting the activation and amplification of pathogenic T cells, particularly in individuals with a genetic predisposition [10,103]. Besides this, most TNF-α inhibitors are immunogenic with consequent anti-drug antibody development and complex immune constitutions [104,105,106]. Anti-TNF-α agents can also impair the normal suppression of auto-reactive B cells, leading to an increased lymphocyte presence, or may provoke cellular apoptosis and the consequent release of self-antigens, leading to the loss of tolerance towards the patient’s cells [35]. Finally, the different molecular structures of the anti-TNF-α agents can lead to divergent effects. In this scenario, new studies should be conducted to extend our clinical knowledge on the pathogenic mechanisms underlying these paradoxical reactions to prevent them and to obtain an overall better management of IBD patients.

Author Contributions

Conceptualisation, L.R.L., A.P. and A.G.; methodology, L.R.L., C.C., I.M. and A.P.; data curation, L.R.L., C.C., I.M., A.G. and A.P.; writing—original draft preparation, L.R.L., C.C., I.M. and A.P.; writing—review and editing, L.R.L., C.C., I.M., A.G. and A.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Papadakis, K.A.; Targan, S.R. Tumor necrosis factor: Biology and therapeutic inhibitors. Gastroenterology 2000, 119, 1148–1157. [Google Scholar] [CrossRef]
  2. Nesbitt, A.; Fossati, G.; Bergin, M.; Stephens, P.; Stephens, S.; Foulkes, R.; Brown, D.; Robinson, M.; Bourne, T. Mechanism of action of certolizumab pegol (CDP870): In vitro comparison with other anti-tumor necrosis factor alpha agents. Inflamm. Bowel Dis. 2007, 13, 1323–1332. [Google Scholar] [CrossRef]
  3. Weaver, A.L. Differentiating the new rheumatoid arthritis biologic therapies. J. Clin. Rheumatol. 2003, 9, 99–114. [Google Scholar] [CrossRef]
  4. Sehgal, R.; Stratman, E.J.; Cutlan, J.E. Biologic Agent-Associated Cutaneous Adverse Events: A Single Center Experience. Clin. Med. Res. 2018, 16, 41–46. [Google Scholar] [CrossRef]
  5. Feuerstein, J.D.; Cheifetz, A.S. Miscellaneous adverse events with biologic agents (excludes infection and malignancy). Gastroenterol. Clin. N. Am. 2014, 43, 543–563. [Google Scholar] [CrossRef]
  6. Lian, N.; Zhang, L.; Chen, M. Tumor necrosis factors-alpha inhibition-induced paradoxical psoriasis: A case series and literature review. Dermatol. Ther. 2020, 33, e14225. [Google Scholar] [CrossRef]
  7. Mylonas, A.; Conrad, C. Psoriasis: Classical vs. Paradoxical. The Yin-Yang of TNF and Type I Interferon. Front. Immunol. 2018, 9, 2746. [Google Scholar] [CrossRef]
  8. Stoffel, E.; Maier, H.; Riedl, E.; Brüggen, M.-C.; Reininger, B.; Schaschinger, M.; Bangert, C.; Guenova, E.; Stingl, G.; Brunner, P. Analysis of anti-tumour necrosis factor-induced skin lesions reveals strong T helper 1 activation with some distinct immunological characteristics. Br. J. Dermatol. 2018, 178, 1151–1162. [Google Scholar] [CrossRef]
  9. Fania, L.; Morelli, M.; Scarponi, C.; Mercurio, L.; Scopelliti, F.; Cattani, C.; Scaglione, G.L.; Tonanzi, T.; Pilla, M.A.; Pagnanelli, G.; et al. Paradoxical psoriasis induced by TNF-alpha blockade shows immunological features typical of the early phase of psoriasis development. J. Pathol. Clin. Res. 2020, 6, 55–68. [Google Scholar] [CrossRef]
  10. Collamer, A.N.; Battafarano, D.F. Psoriatic skin lesions induced by tumor necrosis factor antagonist therapy: Clinical features and possible immunopathogenesis. Semin. Arthritis Rheum. 2010, 40, 233–240. [Google Scholar] [CrossRef]
  11. Bucalo, A.; Rega, F.; Zangrilli, A.; Silvestri, V.; Valentini, V.; Scafetta, G.; Marraffa, F.; Grassi, S.; Rogante, E.; Piccolo, A.; et al. Paradoxical Psoriasis Induced by Anti-TNFalpha Treatment: Evaluation of Disease-Specific Clinical and Genetic Markers. Int. J. Mol. Sci. 2020, 21, 7873. [Google Scholar] [CrossRef]
  12. Weizman, A.V.; Sharma, R.; Afzal, N.M.; Xu, W.; Walsh, S.; Stempak, J.M.; Nguyen, G.C.; Croitoru, K.; Steinhart, A.H.; Silverberg, M.S. Stricturing and Fistulizing Crohn’s Disease Is Associated with Anti-tumor Necrosis Factor-Induced Psoriasis in Patients with Inflammatory Bowel Disease. Dig. Dis. Sci. 2018, 63, 2430–2438. [Google Scholar] [CrossRef]
  13. Almutairi, D.; Sheasgreen, C.; Weizman, A.; Alavi, A. Generalised Pustular Psoriasis Induced by Infliximab in a Patient with Inflammatory Bowel Disease. J. Cutan. Med. Surg. 2018, 22, 507–510. [Google Scholar] [CrossRef]
  14. Courbette, O.; Aupiais, C.; Viala, J.; Hugot, J.-P.; Louveau, B.; Chatenoud, L.; Bourrat, E.; Martinez-Vinson, C. Infliximab Paradoxical Psoriasis in a Cohort of Children with Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2019, 69, 189–193. [Google Scholar] [CrossRef]
  15. Loebenstein, M.; Schulberg, J.D. Anti-TNF Rechallenge After Infliximab Induced Lupus in Crohn’s Disease. Gastroenterology 2020, 158, 2069–2071. [Google Scholar] [CrossRef]
  16. Perez-De-Lis, M.; Retamozo, S.; Flores-Chávez, A.; Kostov, B.; Perez-Alvarez, R.; Brito-Zerón, P.; Ramos-Casals, M. Autoimmune diseases induced by biological agents. A review of 12,731 cases (BIOGEAS Registry). Expert Opin. Drug Saf. 2017, 16, 1255–1271. [Google Scholar] [CrossRef]
  17. Hoffman, B.J. Sensitivity to sulfadiazine resembling acute disseminated lupus erythematosus. Arch. Dermatol. Syph. 1945, 51, 190–192. [Google Scholar] [CrossRef]
  18. Almoallim, H.; Al-Ghamdi, Y.; Almaghrabi, H.; Alyasi, O. Anti-Tumor Necrosis Factor-alpha Induced Systemic Lupus Erythematosus. Open Rheumatol. J. 2012, 6, 315–319. [Google Scholar] [CrossRef]
  19. Jafri, F.; Sammut, A. A rare case of suicidal ideation related to Adalimumab use. Open Access Rheumatol. 2018, 10, 113–115. [Google Scholar] [CrossRef]
  20. Picardo, S.; So, K.; Venugopal, K. Anti-TNF-induced lupus in patients with inflammatory bowel disease. JGH Open 2020, 4, 507–510. [Google Scholar] [CrossRef]
  21. Shovman, O.; Tamar, S.; Amital, H.; Watad, A.; Shoenfeld, Y. Diverse patterns of anti-TNF-alpha-induced lupus: Case series and review of the literature. Clin. Rheumatol. 2018, 37, 563–568. [Google Scholar] [CrossRef]
  22. Turk, D.; Bs, G.V.; Lyons, A.B.; Parks-Miller, A.; Nelson, T.; Meysami, A.; Hamzavi, I.H. Anti-tumor necrosis factor (TNF)-induced lupus in a patient with hidradenitis suppurativa. Int. J. Derm. 2020, 59, e73–e74. [Google Scholar] [CrossRef]
  23. De Bandt, M. Anti-TNF-alpha-induced lupus. Arthritis Res. Ther. 2019, 21, 235. [Google Scholar] [CrossRef]
  24. Tauber, M.; Buche, S.; Reygagne, P.; Berthelot, J.-M.; Aubin, F.; Ghislain, P.-D.; Cohen, J.-D.; Coquerelle, P.; Goujon, E.; Jullien, D.; et al. Alopecia areata occurring during anti-TNF therapy: A national multicenter prospective study. J. Am. Acad. Derm. 2014, 70, 1146–1149. [Google Scholar] [CrossRef]
  25. Youssef, R.; Saad, S.; Hendrick, S. Alopecia universalis and onychodystrophy during treatment with adalimumab. Bayl. Univ. Med. Cent. Proc. 2020, 33, 596–597. [Google Scholar] [CrossRef]
  26. Andrade, P.; Lopes, S.; Albuquerque, A.; Osório, F.; Pardal, J.; Macedo, G. Oral Lichen Planus in IBD Patients: A Paradoxical Adverse Effect of Anti-TNF-alpha Therapy. Dig. Dis. Sci. 2015, 60, 2746–2749. [Google Scholar] [CrossRef]
  27. Oliveira, S.C.; Vasconcelos, A.H.C.; Magalhães, E.P.B.; Corrêa, F.J.V.; Rodrigues, C.E.M. Clinical, Histopathological and Outcome Analysis of Five Patients with Lichenoid Eruption Following Anti-Tumor Necrosis Factor-Alpha Therapy for Ankylosing Spondylitis: Report of One Case and Review of the Literature. Cureus 2020, 12, e10598. [Google Scholar] [CrossRef]
  28. Rashighi, M.; Harris, J.E. Vitiligo Pathogenesis and Emerging Treatments. Derm. Clin. 2017, 35, 257–265. [Google Scholar] [CrossRef]
  29. Attwa, E.; Gamil, H.; Assaf, M.; Ghonemy, S. Over-expression of tumor necrosis factor-alpha in vitiligo lesions after narrow-band UVB therapy: An immunohistochemical study. Arch. Derm. Res. 2012, 304, 823–830. [Google Scholar] [CrossRef]
  30. Kim, N.H.; Torchia, D.; Rouhani, P.; Roberts, B.; Romanelli, P. Tumor necrosis factor-alpha in vitiligo: Direct correlation between tissue levels and clinical parameters. Cutan. Ocul. Toxicol. 2011, 30, 225–227. [Google Scholar] [CrossRef]
  31. Carvalho, C.L.; Ortigosa, L.C. Segmental vitiligo after infliximab use for rheumatoid arthritis—A case report. Bras. Derm. 2014, 89, 154–156. [Google Scholar] [CrossRef] [PubMed]
  32. Bae, J.M.; Kim, M.; Lee, H.H.; Kim, K.-J.; Shin, H.; Ju, H.J.; Kim, G.M.; Park, C.J.; Park, H.J. Increased Risk of Vitiligo Following Anti-Tumor Necrosis Factor Therapy: A 10-Year Population-Based Cohort Study. J. Investig. Derm. 2018, 138, 768–774. [Google Scholar] [CrossRef] [PubMed]
  33. Brunasso, A.M.; Aberer, W.; Massone, C. New onset of dermatomyositis/polymyositis during anti-TNF-alpha therapies: A systematic literature review. Sci. World J. 2014, 2014, 179180. [Google Scholar] [CrossRef] [PubMed]
  34. Takata, M.; Yamasaki, A.; Yamada, N.; Hagino, H.; Funaki, Y.; Harada, T.; Okazaki, R.; Hasegawa, Y.; Fukushima, T.; Morita, M.; et al. A case of clinically amyopathic dermatomyositis that developed during anti-TNF-alpha therapy for rheumatoid arthritis. Allergol. Int 2018, 67, 286–288. [Google Scholar] [CrossRef]
  35. Lopetuso, L.R.; Mocci, G.; Marzo, M.; D’Aversa, F.; Rapaccini, G.L.; Guidi, L.; Armuzzi, A.; Gasbarrini, A.; Papa, A. Harmful Effects and Potential Benefits of Anti-Tumor Necrosis Factor (TNF)-alpha on the Liver. Int. J. Mol. Sci. 2018, 19, 2199. [Google Scholar] [CrossRef]
  36. French, J.B.; Bonacini, M.; Ghabril, M.; Foureau, D.; Bonkovsky, H.L. Hepatotoxicity Associated with the Use of Anti-TNF-alpha Agents. Drug Saf. 2016, 39, 199–208. [Google Scholar] [CrossRef]
  37. Vollmer, O.; Felten, R.; Mertz, P.; Lebrun-Vignes, B.; Salem, J.E.; Arnaud, L. Characterisation of autoimmune hepatitis associated with the use of anti-TNFalpha agents: An analysis of 389 cases in VigiBase. Autoimmun. Rev. 2020, 19, 102460. [Google Scholar] [CrossRef]
  38. Mancini, S.; Amorotti, E.; Vecchio, S.; De Leon, M.P.; Roncucci, L. Infliximab-related hepatitis: Discussion of a case and review of the literature. Intern. Emerg. Med. 2010, 5, 193–200. [Google Scholar] [CrossRef]
  39. Parekh, R.; Kaur, N. Liver Injury Secondary to Anti-TNF-Alpha Therapy in Inflammatory Bowel Disease: A Case Series and Review of the Literature. Case Rep. Gastrointest. Med. 2014, 2014, 956463. [Google Scholar] [CrossRef]
  40. Ierardi, E.; Della Valle, N.; Nacchiero, M.C.; De Francesco, V.; Stoppino, G.; Panella, C. onset of liver damage after a single administration of infliximab in a patient with refractory ulcerative colitis. Clin. Drug Investig. 2006, 26, 673–676. [Google Scholar] [CrossRef]
  41. Tobon, G.J.; Cañas, C.; Jaller, J.-J.; Restrepo, J.-C.; Anaya, J.-M. Serious liver disease induced by infliximab. Clin. Rheumatol. 2007, 26, 578–581. [Google Scholar] [CrossRef] [PubMed]
  42. Colina, F.; Molero, A.; Casís, B.; Martínez-Montiel, P. Infliximab-related hepatitis: A case study and literature review. Dig. Dis. Sci. 2013, 58, 3362–3367. [Google Scholar] [CrossRef]
  43. Bjornsson, E.S.; Gunnarsson, B.I.; Gröndal, G.; Jonasson, J.G.; Einarsdottir, R.; Ludviksson, B.R.; Gudbjörnsson, B.; Olafsson, S. risk of drug-induced liver injury from tumor necrosis factor antagonists. Clin. Gastroenterol. Hepatol. 2015, 13, 602–608. [Google Scholar] [CrossRef] [PubMed]
  44. Haennig, A.; Bonnet, D.; Thebault, S.; Alric, L. Infliximab-induced acute hepatitis during Crohn’s disease therapy: Absence of cross-toxicity with adalimumab. Gastroenterol. Clin. Biol. 2010, 34, e7–e8. [Google Scholar] [CrossRef]
  45. Goldfeld, D.A.; Verna, E.C.; Lefkowitch, J.; Swaminath, A. Infliximab-induced autoimmune hepatitis with successful switch to adalimumab in a patient with Crohn’s disease: The index case. Dig. Dis. Sci. 2011, 56, 3386–3388. [Google Scholar] [CrossRef] [PubMed]
  46. Ghabril, M.; Bonkovsky, H.L.; Kum, C.; Davern, T.; Hayashi, P.H.; Kleiner, D.; Serrano, J.; Rochon, J.; Fontana, R.J.; Bonacini, M. Liver injury from tumor necrosis factor-alpha antagonists: Analysis of thirty-four cases. Clin. Gastroenterol. Hepatol. 2013, 11, 558–564.e3. [Google Scholar] [CrossRef]
  47. Dang, L.J.; Lubel, J.S.; Gunatheesan, S.; Hosking, P.; Su, J. Drug-induced lupus and autoimmune hepatitis secondary to infliximab for psoriasis. Australas. J. Derm. 2014, 55, 75–79. [Google Scholar] [CrossRef]
  48. Cravo, M.; Silva, R.; Serrano, M. Autoimmune hepatitis induced by infliximab in a patient with Crohn’s disease with no relapse after switching to adalimumab. BioDrugs 2010, 24 (Suppl. 1), 25–27. [Google Scholar] [CrossRef] [PubMed]
  49. Chen, J.; Chi, S.; Li, F.; Yang, J.; Cho, W.C.; Liu, X. Biologics-induced interstitial lung diseases in rheumatic patients: Facts and controversies. Expert Opin. Biol. 2017, 17, 265–283. [Google Scholar] [CrossRef] [PubMed]
  50. Panopoulos, S.T.; Sfikakis, P.P. Biological treatments and connective tissue disease associated interstitial lung disease. Curr. Opin. Pulm. Med. 2011, 17, 362–367. [Google Scholar] [CrossRef]
  51. Sharp, C.; Dodds, N.; Mayers, L.; Millar, A.; Gunawardena, H.; Adamali, H. The role of biologics in treatment of connective tissue disease-associated interstitial lung disease. QJM Int. J. Med. 2015, 108, 683–688. [Google Scholar] [CrossRef] [PubMed]
  52. Dias, O.M.; Pereira, D.A.S.; Baldi, B.; Costa, A.N.; Athanazio, R.; Kairalla, R.; Carvalho, C. Adalimumab-induced acute interstitial lung disease in a patient with rheumatoid arthritis. J. Bras. Pneumol. 2014, 40, 77–81. [Google Scholar] [CrossRef]
  53. Atzeni, F.; Boiardi, L.; Salli, S.; Benucci, M.; Sarzi-Puttini, P. Lung involvement and drug-induced lung disease in patients with rheumatoid arthritis. Expert Rev. Clin. Immunol. 2013, 9, 649–657. [Google Scholar] [CrossRef] [PubMed]
  54. Ramos-Casals, M.; Perez-Alvarez, R.; Perez-De-Lis, M.; Xaubet, A.; Bosch, X. Pulmonary disorders induced by monoclonal antibodies in patients with rheumatologic autoimmune diseases. Am. J. Med. 2011, 124, 386–394. [Google Scholar] [CrossRef] [PubMed]
  55. Perez-Alvarez, R.; Perez-De-Lis, M.; Diaz-Lagares, C.; Pego-Reigosa, J.M.; Retamozo, S.; Bove, A.; Brito-Zeron, P.; Bosch, X.; Ramos-Casals, M. Interstitial lung disease induced or exacerbated by TNF-targeted therapies: Analysis of 122 cases. Semin. Arthritis Rheum. 2011, 41, 256–264. [Google Scholar] [CrossRef] [PubMed]
  56. Roubille, C.; Haraoui, B. Interstitial lung diseases induced or exacerbated by DMARDS and biologic agents in rheumatoid arthritis: A systematic literature review. Semin. Arthritis Rheum. 2014, 43, 613–626. [Google Scholar] [CrossRef]
  57. Komiya, K.; Ishii, H.; Fujita, N.; Oka, H.; Iwata, A.; Sonoda, H.; Kadota, J.-I. Adalimumab-induced interstitial pneumonia with an improvement of pre-existing rheumatoid arthritis-associated lung involvement. Intern. Med. 2011, 50, 749–751. [Google Scholar] [CrossRef]
  58. Allanore, Y.; Devos-François, G.; Caramella, C.; Boumier, P.; Jounieaux, V.; Kahan, A. Fatal exacerbation of fibrosing alveolitis associated with systemic sclerosis in a patient treated with adalimumab. Ann. Rheum. Dis. 2006, 65, 834–835. [Google Scholar] [CrossRef]
  59. Sen, S.; Jordan, K.; Boes, T.J.; Peltz, C. Infliximab-induced non-specific interstitial pneumonia. Am. J. Med. Sci. 2012, 344, 75–78. [Google Scholar] [CrossRef]
  60. Rofaiel, R.; Kohli, S.; Mura, M.; Hosseini-Moghaddam, S.M. A 53-year-old man with dyspnoea, respiratory failure, consistent with infliximab-induced acute interstitial pneumonitis after an accelerated induction dosing schedule. BMJ Case Rep. 2017, 2017, bcr-2017. [Google Scholar] [CrossRef]
  61. Herrinton, L.J.; Harrold, L.R.; Liu, L.; Raebel, M.A.; Taharka, A.; Winthrop, K.L.; Solomon, D.H.; Curtis, J.R.; Lewis, J.D.; Saag, K.G. Association between anti-TNF-alpha therapy and interstitial lung disease. Pharmacoepidemiol. Drug Saf. 2013, 22, 394–402. [Google Scholar] [CrossRef]
  62. Nakashita, T.; Ando, K.; Kaneko, N.; Takahashi, K.; Motojima, S. Potential risk of TNF inhibitors on the progression of interstitial lung disease in patients with rheumatoid arthritis. BMJ Open 2014, 4, e005615. [Google Scholar] [CrossRef]
  63. Huang, Y.; Lin, W.; Chen, Z.; Wang, Y.; Huang, Y.; Tu, S. Effect of tumor necrosis factor inhibitors on interstitial lung disease in rheumatoid arthritis: Angel or demon? Drug Des. Devel. 2019, 13, 2111–2125. [Google Scholar] [CrossRef]
  64. Miyazaki, Y.; Araki, K.; Vesin, C.; Garcia, I.; Kapanci, Y.; Whitsett, J.A.; Piguet, P.F.; Vassalli, P. Expression of a tumor necrosis factor-alpha transgene in murine lung causes lymphocytic and fibrosing alveolitis. A mouse model of progressive pulmonary fibrosis. J. Clin. Investig. 1995, 96, 250–259. [Google Scholar] [CrossRef]
  65. Sullivan, D.E.; Ferris, M.; Nguyen, H.; Abboud, E.; Brody, A.R. TNF-alpha induces TGF-beta1 expression in lung fibroblasts at the transcriptional level via AP-1 activation. J. Cell Mol. Med. 2009, 13, 1866–1876. [Google Scholar] [CrossRef]
  66. Kuroki, M.; Noguchi, Y.; Shimono, M.; Tomono, K.; Tashiro, T.; Obata, Y.; Nakayama, E.; Kohno, S. Repression of bleomycin-induced pneumopathy by TNF. J. Immunol. 2003, 170, 567–574. [Google Scholar] [CrossRef]
  67. Piga, M.; Chessa, E.; Ibba, V.; Mura, V.; Floris, A.; Cauli, A.; Mathieu, A. Biologics-induced autoimmune renal disorders in chronic inflammatory rheumatic diseases: Systematic literature review and analysis of a monocentric cohort. Autoimmun. Rev. 2014, 13, 873–879. [Google Scholar] [CrossRef]
  68. Simms, R.; Kipgen, D.; Dahill, S.; Marshall, D.; Rodger, R.S. ANCA-associated renal vasculitis following anti-tumor necrosis factor alpha therapy. Am. J. Kidney Dis. 2008, 51, e11–e14. [Google Scholar] [CrossRef]
  69. Ashok, D.; Dubey, S.; Tomlinson, I. C-ANCA positive systemic vasculitis in a patient with rheumatoid arthritis treated with infliximab. Clin. Rheumatol. 2008, 27, 261–264. [Google Scholar] [CrossRef]
  70. Lee, A.; Kasama, R.; Evangelisto, A.; Elfenbein, B.; Falasca, G. Henoch-Schonlein purpura after etanercept therapy for psoriasis. J. Clin. Rheumatol. 2006, 12, 249–251. [Google Scholar] [CrossRef]
  71. Yahya, T.M.; Dhanyamraju, S.; Harrington, T.M.; Prichard, J.W. Spontaneous resolution of lupus nephritis following withdrawal of etanercept. Ann. Clin. Lab. Sci. 2013, 43, 447–449. [Google Scholar] [PubMed]
  72. den Broeder, A.A.; Assmann, K.J.; Van Riel, P.L.; Wetzels, J.F. Nephrotic syndrome as a complication of anti-TNFalpha in a patient with rheumatoid arthritis. Neth. J. Med. 2003, 61, 137–141. [Google Scholar]
  73. Castro Corredor, D.; de la Nieta, M.D.S.; de Lara Simon, I.M. Acute tubulointerstitial nephritis in an HLA-B27-positive patient with axial spondyloarthritis being treated with adalimumab. Reum. Clin. 2019, 15, 179–181. [Google Scholar] [CrossRef]
  74. Korsten, P.; Sweiss, N.J.; Nagorsnik, U.; Niewold, T.B.; Gröne, H.-J.; Gross, O.; Müller, G.A. Drug-induced granulomatous interstitial nephritis in a patient with ankylosing spondylitis during therapy with adalimumab. Am. J. Kidney Dis. 2010, 56, e17–e21. [Google Scholar] [CrossRef] [PubMed]
  75. Akiyama, M.; Kaneko, Y.; Hanaoka, H.; Kuwana, M.; Takeuchi, T. Acute kidney injury due to renal sarcoidosis during etanercept therapy: A case report and literature review. Intern. Med. 2015, 54, 1131–1134. [Google Scholar] [CrossRef] [PubMed]
  76. Sokumbi, O.; Wetter, D.A.; Makol, A.; Warrington, K.J. vasculitis associated with tumor necrosis factor-alpha inhibitors. Mayo. Clin. Proc. 2012, 87, 739–745. [Google Scholar] [CrossRef]
  77. Saint Marcoux, B.; De Bandt, M. on behalf of the CRI (Club Rhumatismes et Inflammation). Vasculitides induced by TNFalpha antagonists: A study in 39 patients in France . Jt. Bone Spine 2006, 73, 710–713. [Google Scholar]
  78. Ramos-Casals, M.; Brito-Zerón, P.; Muñoz, S.; Soria, N.; Galiana, D.; Bertolaccini, L.; Cuadrado, M.-J.; Khamashta, M.A. Autoimmune diseases induced by TNF-targeted therapies: Analysis of 233 cases. Medicine 2007, 86, 242–251. [Google Scholar] [CrossRef] [PubMed]
  79. Moustou, A.E.; Matekovits, A.; Dessinioti, C.; Antoniou, C.; Sfikakis, P.P.; Stratigos, A.J. Cutaneous side effects of anti-tumor necrosis factor biologic therapy: A clinical review. J. Am. Acad. Derm. 2009, 61, 486–504. [Google Scholar] [CrossRef]
  80. Kemanetzoglou, E.; Andreadou, E. CNS Demyelination with TNF-alpha Blockers. Curr. Neurol. Neurosci. Rep. 2017, 17, 36. [Google Scholar] [CrossRef]
  81. Deepak, P.; Stobaugh, D.J.; Sherid, M.; Sifuentes, H.; Ehrenpreis, E.D. Neurological events with tumour necrosis factor alpha inhibitors reported to the Food and Drug Administration Adverse Event Reporting System. Aliment. Pharm. 2013, 38, 388–396. [Google Scholar] [CrossRef]
  82. Cruz Fernandez-Espartero, M.; Pérez-Zafrilla, B.; Naranjo, A.; Esteban, C.; Ortiz, A.M.; Gómez-Reino, J.J.; Carmona, L. Demyelinating disease in patients treated with TNF antagonists in rheumatology: Data from BIOBADASER, a pharmacovigilance database, and a systematic review. Semin. Arthritis Rheum. 2011, 41, 524–533. [Google Scholar] [CrossRef]
  83. Daïen, C.I.; Monnier, A.; Claudepierre, P.; Constantin, A.; Eschard, J.-P.; Houvenagel, E.; Samimi, M.; Pavy, S.; Pertuiset, E.; Toussirot, E.; et al. Sarcoid-like granulomatosis in patients treated with tumor necrosis factor blockers: 10 cases. Rheumatology 2009, 48, 883–886. [Google Scholar] [CrossRef]
  84. Burns, A.M.; Green, P.J.; Pasternak, S. Etanercept-induced cutaneous and pulmonary sarcoid-like granulomas resolving with adalimumab. J. Cutan. Pathol. 2012, 39, 289–293. [Google Scholar] [CrossRef]
  85. Sim, J.K.; Lee, S.Y.; Shim, J.J.; Kang, K.H. Pulmonary Sarcoidosis Induced by Adalimumab: A Case Report and Literature Review. Yonsei Med. J. 2016, 57, 272–273. [Google Scholar] [CrossRef]
  86. Decock, A.; Van Assche, G.; Vermeire, S.; Wuyts, W.; Ferrante, M. Sarcoidosis-Like Lesions: Another Paradoxical Reaction to Anti-TNF Therapy? J. Crohns. Colitis. 2017, 11, 378–383. [Google Scholar] [CrossRef]
  87. Fernandez-Trujillo, L.; Iriarte, M.B.; Puerta, G.; Morales, E.I.; Sua, L.F.; Cañas, C.A. Early instauration granulomatous pneumonitis associated with use of etanercept in seronegative spondyloarthropathy: Case report. Respir. Med. Case Rep. 2020, 30, 101079. [Google Scholar] [CrossRef]
  88. Park, S.K.; Hwang, P.-H.; Yun, S.-K.; Kim, H.-U.; Park, J. Tumor Necrosis Factor Alpha Blocker-Induced Erythrodermic Sarcoidosis in with Juvenile Rheumatoid Arthritis: A Case Report and Review of the Literature. Ann. Derm. 2017, 29, 74–78. [Google Scholar] [CrossRef]
  89. Isshiki, T.; Homma, S.; Eishi, Y.; Yabe, M.; Koyama, K.; Nishioka, Y.; Yamaguchi, T.; Uchida, K.; Yamamoto, K.; Ohashi, K.; et al. Immunohistochemical Detection of Propionibacterium acnes in Granulomas for Differentiating Sarcoidosis from Other Granulomatous Diseases Utilising an Automated System with a Commercially Available PAB Antibody. Microorganisms 2021, 9, 1668. [Google Scholar] [CrossRef]
  90. Mengi, G.; Gogus, F. A Rare Adverse Effect of Anti-Tumor Necrosis Factor Alpha Therapy: Sarcoidosis. Arch. Rheumatol. 2017, 32, 67–70. [Google Scholar] [CrossRef]
  91. Unterstell, N.; Bressan, A.L.; Serpa, L.A.; Castro, P.P.D.F.E.; Gripp, A.C. Systemic sarcoidosis induced by etanercept: First Brazilian case report. Bras. Derm. 2013, 88 (Suppl. 1), 197–199. [Google Scholar] [CrossRef] [PubMed]
  92. Jung, J.H.; Kim, J.H.; Song, G.G. Adalimumab-induced pulmonary sarcoidosis not progressing upon treatment with etanercept. Z. Rheumatol. 2017, 76, 372–374. [Google Scholar] [CrossRef] [PubMed]
  93. Okoshi, M.; Sato, H.; Honma, T.; Terai, S. Rare paradoxical adverse event in Crohn’s disease: A case report. Ann. Transl. Med. 2020, 8, 133. [Google Scholar] [CrossRef] [PubMed]
  94. Massara, A.; Cavazzini, L.; La Corte, R.; Trotta, F. Sarcoidosis appearing during anti-tumor necrosis factor alpha therapy: A new “class effect” paradoxical phenomenon. Two case reports and literature review. Semin. Arthritis Rheum. 2010, 39, 313–319. [Google Scholar] [CrossRef]
  95. O’Toole, A.; Lucci, M.; Korzenik, J. Inflammatory Bowel Disease Provoked by Etanercept: Report of 443 Possible Cases Combined from an IBD Referral Center and the FDA. Dig. Dis. Sci. 2016, 61, 1772–1774. [Google Scholar] [CrossRef]
  96. Tolu, S.; Rezvani, A.; Hindioglu, N.; Korkmaz, M.C. Etanercept-induced Crohn’s disease in ankylosing spondylitis: A case report and review of the literature. Rheumatol. Int. 2018, 38, 2157–2162. [Google Scholar] [CrossRef]
  97. Sandborn, W.J.; Hanauer, S.B.; Katz, S.; Safdi, M.; Wolf, D.G.; Baerg, R.D.; Tremaine, W.J.; Johnson, T.; Diehl, N.N.; Zinsmeister, A.R. Etanercept for active Crohn’s disease: A randomised, double-blind, placebo-controlled trial. Gastroenterology 2001, 121, 1088–1094. [Google Scholar] [CrossRef]
  98. van Dijken, T.D.; Vastert, S.J.; Gerloni, V.M.; Pontikaki, I.; Linnemann, K.; Girschick, H.; Armbrust, W.; Minden, K.; Prince, F.H.M.; Kokke, F.T.M.; et al. Development of inflammatory bowel disease in patients with juvenile idiopathic arthritis treated with etanercept. J. Rheumatol. 2011, 38, 1441–1446. [Google Scholar] [CrossRef]
  99. Barthel, D.; Ganser, G.; Kuester, R.-M.; Onken, N.; Minden, K.; Girschick, H.J.; Hospach, A.; Horneff, G. Inflammatory Bowel Disease in Juvenile Idiopathic Arthritis Patients Treated with Biologics. J. Rheumatol. 2015, 42, 2160–2165. [Google Scholar] [CrossRef]
  100. Bieber, A.; Fawaz, A.; Novofastovski, I.; Mader, R. Anti-tumor Necrosis Factor-alpha Therapy Associated with Inflammatory Bowel Disease: Three Cases and a Systematic Literature Review. J. Rheumatol. 2017, 44, 1088–1095. [Google Scholar] [CrossRef]
  101. Forouzandeh, M.; Vazquez, T.; Nouri, K.; Forouzandeh, B. The Paradoxical Induction of Crohn’s Disease Following Treatment of Psoriatic Arthritis with Etanercept. J. Drugs Derm. 2019, 18, 832–834. [Google Scholar]
  102. Kolios, A.G.A.; Biedermann, L.; Weber, A.; Navarini, A.A.; Meier, J.; Cozzio, A.; French, L.E. Paradoxical ulcerative colitis during adalimumab treatment of psoriasis resolved by switch to ustekinumab. Br. J. Derm. 2018, 178, 551–555. [Google Scholar] [CrossRef] [PubMed]
  103. Palucka, A.K.; Blanck, J.-P.; Bennett, L.; Pascual, V.; Banchereau, J. Cross-regulation of TNF and IFN-alpha in autoimmune diseases. Proc. Natl. Acad. Sci. USA 2005, 102, 3372–3377. [Google Scholar] [CrossRef] [PubMed]
  104. Jani, M.; Dixon, W.G.; Chinoy, H. Drug safety and immunogenicity of tumour necrosis factor inhibitors: The story so far. Rheumatology 2018, 57, 1896–1907. [Google Scholar] [CrossRef] [PubMed]
  105. Dalle Vedove, C.; Simon, J.C.; Girolomoni, G. Drug-induced lupus erythematosus with emphasis on skin manifestations and the role of anti-TNFalpha agents. J. Dtsch. Derm. Ges. 2012, 10, 889–897. [Google Scholar]
  106. Guillevin, L.; Mouthon, L. Tumor necrosis factor-alpha blockade and the risk of vasculitis. J. Rheumatol. 2004, 31, 1885–1887. [Google Scholar] [PubMed]
Table 1. Autoimmune clinical manifestations caused by anti-TNF-α agents.
Table 1. Autoimmune clinical manifestations caused by anti-TNF-α agents.
Autoimmune
Clinical Manifestation
Most Involved
Anti-TNF-α
Most Involved Disease/
Type of Patients
TreatmentSafe Switch to Alternative Anti-TNF-α Reported
PsoriasisIFXRAStop Anti-TNF-α
Anti-TNF-induced LupusIFX-Stop Anti-TNF-α
Vitiligo and AAETAYoung patientsStop Anti-TNF
Autoimmune HepatitisIFX-Stop Anti-TNF-α ± steroids
Interstitial Lung DiseasesIFX and ETARAStop Anti-TNF-α
Renal DiseasesETARAStop Anti-TNF
Vasculitis\-Stop Anti-TNF-α ± steroids
DemyelinationETARAStop Anti-TNF-α ± steroids
SarcoidosisETARAStop Anti-TNF-α ± steroids
New-onset IBDETAASStop Anti-TNF-α
Abbreviations: TNF-α: tumour necrosis factor-α; IFX: infliximab; RA: rheumatoid arthritis; ETA: etanercept; AA: alopecia areata; AS: ankylosing spondylitis.
Table 2. Pathophysiology of paradoxical reactions and related clinical manifestations.
Table 2. Pathophysiology of paradoxical reactions and related clinical manifestations.
Primary Mechanism Involved in Paradoxical ReactionsClinical Manifestation
Overexpression of IFN-γ and homing of Th1 cellsPsoriasis and other cutaneous manifestations (alopecia areata, lichenoid eruption, and vitiligo) and new-onset IBD
Activation of IL-23/Th-17 axisPsoriasis and sarcoidosis
Anti-drug antibodies formationAnti-TNF-induced lupus, systemic and cutaneous vasculitis, interstitial lung disease, and glomerulonephritis
Blockade of soluble TNF-α by ETA Sarcoidosis, new-onset IBD, and demyelination
Decrease of TNFR2 receptorsDemyelination
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lopetuso, L.R.; Cuomo, C.; Mignini, I.; Gasbarrini, A.; Papa, A. Focus on Anti-Tumour Necrosis Factor (TNF)-α-Related Autoimmune Diseases. Int. J. Mol. Sci. 2023, 24, 8187. https://doi.org/10.3390/ijms24098187

AMA Style

Lopetuso LR, Cuomo C, Mignini I, Gasbarrini A, Papa A. Focus on Anti-Tumour Necrosis Factor (TNF)-α-Related Autoimmune Diseases. International Journal of Molecular Sciences. 2023; 24(9):8187. https://doi.org/10.3390/ijms24098187

Chicago/Turabian Style

Lopetuso, Loris Riccardo, Claudia Cuomo, Irene Mignini, Antonio Gasbarrini, and Alfredo Papa. 2023. "Focus on Anti-Tumour Necrosis Factor (TNF)-α-Related Autoimmune Diseases" International Journal of Molecular Sciences 24, no. 9: 8187. https://doi.org/10.3390/ijms24098187

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop