Next Article in Journal
Clinical Nomogram Model for Pre-Operative Prediction of Microvascular Invasion of Hepatocellular Carcinoma before Hepatectomy
Previous Article in Journal
Disease Activity, Inflammation Markers, and Quality of Life Are Associated with Muscle Strength in Croatian Rheumatoid Arthritis Patients—A National-Based Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neurological Side Effects of TNF-α Inhibitors Revisited: A Review of Case Reports

1
Faculty of Medicine, Victor Babes University of Medicine and Pharmacy, 2 Eftimie Murgu, 300041 Timisoara, Romania
2
Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, 2 Eftimie Murgu, 300041 Timisoara, Romania
*
Author to whom correspondence should be addressed.
Medicina 2024, 60(9), 1409; https://doi.org/10.3390/medicina60091409
Submission received: 8 August 2024 / Revised: 27 August 2024 / Accepted: 28 August 2024 / Published: 28 August 2024
(This article belongs to the Section Neurology)

Abstract

:
Over the past two decades, the use of tumor necrosis factor alpha (TNF-α) inhibitors has significantly improved the treatment of patients with immune-mediated inflammatory diseases. Firstly, introduced for rheumatoid arthritis, these inhibitors are currently approved and used for a variety of conditions, including ankylosing spondylitis, Crohn’s disease, juvenile idiopathic arthritis, psoriasis, psoriatic arthritis, ulcerative colitis, and chronic uveitis. Despite their immense therapeutic efficacy, TNF-α inhibitors have been associated with neurological adverse effects that bring new clinical challenges. The present review collects data from multiple studies to evaluate the incidence and the relationship between TNF-α inhibitors and neurological side effects and to explore the potential underlying mechanisms of this association. Moreover, it highlights the importance of patient selection, particularly in the case of individuals with a history of demyelinating diseases, raises awareness for clinicians, and calls for ongoing research that will improve TNF-α targeting strategies and offer safer and more effective therapeutic options.

1. Introduction

Although the treatment of immune-mediated inflammatory diseases is constantly improving, tumor necrosis factor alpha (TNF-α) inhibitors still represent the main therapeutic option, able to significantly alter the physical course of various pathologies. TNF-α inhibitors were introduced in therapy two decades ago as treatment against rheumatoid arthritis, but since then new indications have occurred; currently, TNF-α inhibitors are approved for use in ankylosing spondylitis (AS) [1], Crohn’s disease (CD) [2], juvenile idiopathic arthritis (JIA) [3], psoriasis (PSO) [4], psoriatic arthritis (PsA) [5], rheumatoid arthritis (RA) [6], ulcerative colitis (UC) [7], chronic uveitis [8], and others.
Inhibitory effects against TNF-α were revealed for several molecules both natural and synthetic; an excellent review published in 2022 assessed the main natural compounds that may act as TNF-α inhibitors [9]. The authors introduced several alkaloids, flavonoids, terpenes, and polyphenols as potential TNF-α inhibitors and discussed their interactions with aminoacidic residues, which appear to be essential for their biologic activity; such data may provide future scaffolds for anti-inflammatory drugs. Among synthetic molecules, thalidomide was revealed as active against TNF-α through various mechanisms [10]; its derivatives, lenalidomide [11] and pomalidomide [12] also show TNF-α inhibitor effects that may be exploited in severe pathologies, including malignancies. Other molecules such as xanthine derivatives [13] or bupropion [14] were also identified with TNF-α inhibitor activity. However, most TNF-α inhibitors are monoclonal antibodies, with four such compounds being currently marketed—infliximab, adalimumab, golimumab, and certolizumab (Table 1); in addition, the fifth compound currently approved is etanercept, a fusion protein produced by the recombinant DNA technique. Biological therapy with TNF-α inhibitors has become more affordable once biosimilars were introduced; currently, the five marketed TNF-α inhibitors are on the WHO’s list of essential medicines. Research is ongoing on finding biomarkers that would allow better monitoring of the biological treatment; in addition, new delivery systems with TNF-α inhibitors may be designed in the future with or without associated agents.
Generally, TNF-α inhibitors are considered to display safe pharmacological profiles, being assessed even during pregnancy, where they cross the placenta starting from the 22nd gestational week [15], as well as during the COVID-19 epidemic, where patients under treatment with TNF-α inhibitors exhibited lower rates of morbidity and mortality related to the viral infection compared to the general population [16]. The most common reported adverse effects consist of headaches, rash, anemia, cough, diarrhea, and abdominal discomfort [17]. However, severe adverse reactions have also occurred, such as infections, including tuberculosis, and malignancies [18]. An excellent review with meta-analysis was published in 2021 by Li et al., who emphasized severe side effects occurred during treatment with TNF-α inhibitors; briefly, the meta-analysis suggested an increased risk of infectious events and various malignancies [18]. Controversially, another study, although smaller, found only an increased risk of lymphoma in inflammatory bowel disease patients under TNF-α inhibitor treatment [19]. Systemic lupus erythematosus (SLE) was reported as induced by biologic therapy of psoriasis, particularly in patients with psoriatic arthritis or with comorbidities such as rheumatoid arthritis [20]. The authors reported that the induced SLE can be correlated with the depletion of cytokines IL-2 and IL-10 and an overall cytokine imbalance; however, they concluded that SLE induction is drug-specific and less class-specific, therefore recommending a detailed examination of the patient in order to establish the potential of drug-induced SLE and the specific drug that may trigger the disease. Additionally, prolonged use of TNF-α inhibitors may cause in some patients the development of anti-drug antibodies (i.e., adalimumab), thus promoting adverse effects and reducing drug efficacy; other studies revealed that induced autoantibodies, clinically manifest autoimmune diseases, and paradoxical inflammation may also occur as a result of administered TNF-α inhibitors [21].
In terms of neurological side effects, the literature reports a limited number of cases where such effects appeared, but some consisted of dramatic symptomatology or even the triggering of life-threatening pathologies. This review aims to summarize the most significant data on the neurological damage that may be caused by the administration of TNF-α inhibitors; it will also attempt to distinguish between cases where neurological reactions occurred as a result of the biological treatment and those where they can be regarded as a simultaneous separate condition.
Table 1. Anti-TNF-α agents.
Table 1. Anti-TNF-α agents.
TypeMechanism of ActionDate of Approval (FDA)Approved IndicationsAdministrationPharmacodynamicsRef.
EtanerceptDimeric soluble fusion protein—the extracellular ligand-binding portion of the human p75 TNFR (TNFR2) linked to the Fc portion of human IgG1Inhibits the binding of TNF-α and TNF-β to cell surface TNFRs → renders TNF biologically inactive.1998RA, JIA, PsA, PsO, AS, nr-axSpASubcutaneously weekly
  • Modulates the biological response/decrease in serum levels of E-selectin, ICAM-1, VCAM-1, VEGF, IL-6, IL-1
  • Down-regulates the serum levels of MMP-1 and MMP-3
[22,23,24,25,26]
InfliximabChimeric IG1κ monoclonal antibody (75% human and 25% mouse)Selectively neutralizes the biological activity of TNFα by binding to the tmTNF-α
+ sTNF-α → inhibits the binding of TNFα to TNFR.
No binding/inactivation of TNF-β
1998AS, CD, JIA, PsO, PsA, RA, UC, nr-axSpAIntravenously every 6–8 weeks
  • Decreases serum levels of IL-6 and CRP
  • Reduced the T-cell number in blood vessels + synovium + psoriatic skin lesions
  • Reduced the macrophages in the synovium
  • Modulates the biological response/decrease in serum levels of E-selectin, ICAM-1, VCAM-1, IL-8, MCP-1
[27,28,29,30,31]
AdalimumabFully
human IgG1 monoclonal antibody specific for human TNF-α
Binds specifically to TNF-α → blocks its interaction with the p55 and p75 cell surface TNFR (TNFR1 and TNFR2).
In the presence of complement, lyses cells that express at the surface TNF.
No binding/inactivation of TNF-β
2002AS, CD, JIA, PsO, PsA, RA, UC, Uveitis, nr-axSpASubcutaneously every other week
  • Decreases the expression of adhesion molecules ELAM-1, VCAM-1, ICAM-1, IL-10, IL-12
  • Decreases the serum levels of MMP-1 and MMP-3
  • Decreases the serum levels of CRP, ESR
[32,33,34,35,36,37,38,39,40]
GolimumabFully human IgG1 κ monoclonal antibody specific for human TNF-αSelectively neutralizes the biological activity of TNFα by binding to the tmTNF-α
+ sTNF-α → inhibits the binding of TNFα to TNFR.
No binding/inactivation of TNF-β.
2009AS, PsA, RA, UC, nr-axSpASubcutaneously every month
  • Decreases the serum levels of CRP, IL-6, MMP-3, haptoglobin, ferritin
[41,42,43,44]
Certolizumab pegolHumanized antibody Fab’ fragment conjugated with two polyethylene glycol molecules, specific for human TNF-αSelectively neutralizes the biological activity of TNFα by binding to the tmTNF-α
+ sTNF-α → inhibits the binding of TNFα to TNFR.
No binding/inactivation of TNF-β.
Does not contain a fragment-crystallizable (Fc) region = does not fix complement or cause antibody-dependent cell-mediated cytotoxicity in vitro.
2008AS, CD, PsA, RA, nr-axSpASubcutaneously; remission induction weeks 0, 2 and 4, maintenance of remission every 4 weeks
  • Decreases the serum levels of CRP
[45,46,47]
FDA = U.S. Food and Drug Administration; RA = rheumatoid arthritis, JIA = juvenile idiopathic arthritis, PsA = psoriatic arthritis, PsO = plaque psoriasis, AS = ankylosing spondylitis, CD = Crohn’s disease, UC = ulcerative colitis; ICAM-1 = intercellular adhesion molecule-1; VCAM-1 = vascular cell adhesion molecule-1; MCP-1 = monocyte chemotactic protein-1; IL = interleukin; MMP = matrix metalloproteinase; ELAM = endothelial leucocyte adhesion molecule-1 ; VEGF = vascular endothelial growth factor; ESR = erythrocyte sedimentation rate; nr-axSpA = non-radiographic axial spondyloarthritis.

2. TNF-α Mechanism of Action

TNF-α is produced in many cells but mostly in activated macrophages, T cells, NK cells, endothelial cells, and fibroblasts [48]. The membrane-bound pro-TNF-α, also called transmembrane TNF-α (tmTNF-α), is proteolyzed by a metalloproteinase, TNF-converting enzyme (TECA), in soluble TNF-α (sTNF-α). Both tmTNF-α and sTNF-α can bind to the TNF receptors type 1 and 2 (TNFR1 and TNFR2) and activate consecutively several signaling pathways. Even if at a low concentration, TNFR1 is found in almost all cell types, while TNFR2 is found manly in immune cells [49].
Activation of TNFR1 triggers the interaction with TNFR1-associated death domain (TRADD) and consecutive binding to other proteins, such as TNFR-associated factor 2 (TRAF2), receptor-interacting serine/threonine-protein kinase 1 (RIPK1), transforming growth factor beta-activated kinase 1 (TAK1) and the adapter proteins TAB1 and TAB2, linear ubiquitin chain assembly complex (LUBAC), and cellular inhibitor of apoptosis protein 1 and 2 (cIAP1/2) (Figure 1). Using the ubiquitin ligase E3, C-IAP1/2 ubiquitinate RIPK1 and themselves, enabling the binding of LUBAC, which continues to add linear ubiquitin chains. NEMO will then bind to the linear chains, enabling the recruitment of IKKα/β, which leads to IκB ubiquitination and degradation and consecutive activation of the canonical NF-κB pathway [50]. TAB2/3 (TAK1-binding proteins 2 and 3) transport TAK1 to the complex, thus contributing also to the activation of the MAPK signaling pathway and activator protein-1 (AP-1) [50]. These events will lead to the activation of Nf-κB and to subsequent cell survival, inflammation, defense against pathogens, angiogenesis, methoastasis, and tissue degeneration [51,52]. By removing the polyubiquitin chains, RIPK1 can dissociate from TNFR1 and can bind to RIPK3, leading to the formation of a microfilament-like complex called the necrosome; this aggregate phosphorylates the pseudokinase mixed-lineage kinase domain-like (MLKL) and ultimately induces necroptosis [53]. With time, the activated TNFR1 may be endocytosed; in the cytoplasm, the free receptor can form other complexes with TRADD, Fas-associated protein with death domain (FADD), RIPK1, TRAF2, cIAP1/2, and pro-Caspase-8 [54]. Pro-Caspase-8 is proteolytically cleaved and releases active caspase-8, which in turn activates caspase-3 and -7 and triggers apoptosis.
The TNFR2 receptor does not present a death domain (DD); however, it has a short amino acid sequence that permits the recruitment of adapter protein TRAF1, TRAF2, TRAF 3, and cIAP1/2 (Figure 1). Compared to TNFR1, where its activation led to the canonical activation of the Nf-κB pathway, TNFR2 activates the alternative NF-κB pathway by recruiting the TRAF1/2/3-cIAP1/2 complexes [54]. Specifically, the induced depletion of these cytosolic complexes results in the accumulation of active NF-κB-inducing kinase (NIK), followed by NIK-mediated phosphorylation of ΙΚΚα and protein p100; the phosphorylated p100 is cleaved into p52, which is then translocated to the nucleus, where it regulates gene transcription [55,56]. Activation of the TNFR2 receptor can also trigger the activation of the PI3K/AKT pathway. As a result, AKT can activate the NF-κB pathway and also increase the phosphorylation of STAT5 [57]. Moreover, recent studies demonstrated that TNFR2 activates the JNK pathway in a TRAF2-independent fashion, leading to the phosphorylation of C-Jun [58].

3. Research Methods

The data on the neurological adverse effects associated with tumor necrosis factor alpha (TNF-α) inhibitors was obtained after a thorough literature search on PubMed using specific keywords related to the topic: “neurologic”, “TNF-α inhibitors”, “inflammatory”, and “neurodegenerative”. The search included the entire period of TNF-α inhibitor clinical use (1998—present), starting with the approval by the US Food and Drug Administration (FDA) in 1998 of the first marketed compound, infliximab, for the treatment of Crohn’s disease. Only case reports that provided detailed clinical insights and specific instances of neurological adverse effects linked to TNF-α inhibitor therapy were selected. The literature search identified 113 case reports, which were included in the review, statistically assessed, and discussed.

4. Results

Detailed results are presented in Table 2, where data show that 113 cases of neurologic events were identified, with a mean age of 45; 61% of the patients were women and 39% men. The neurological adverse effects occurred either immediately after the first drug infusion or after various periods of time, up to 10 years of drug usage. The most common primary disease involved was rheumatoid arthritis (31.86%), followed by psoriasis (20.35%) and Crohn’s disease (19.47%) (Figure 2); however, a variety of other autoimmune conditions were also the causes for TNF-α inhibitor administration. Infliximab and adalimumab were the most commonly used, each counting for more than 35% of cases; the second most common was etanercept, while the fewest cases were recorded for certolizumab pegol and golimumab, which were introduced later on the pharmaceutical market. No family or personal history of neurologic conditions was reported in 20% of cases, but this result carries poor significance since more than 70% of the cases did not report any investigation in this regard.
Neurological events occurred presumably in relation to TNF-α therapy. These events consisted of both demyelinating and non-demyelinating conditions; however, the demyelination reactions prevailed (83% versus 17%). INFL shows a strong association with demyelination, more precisely, with CNS (43%) and peripheral (18%) demyelination; INFL also induced neuropathies (15%), neurodegenerative diseases (7.5%), inflammatory demyelination (6.5%), seizures (5%), and neurosarcoidosis (5%) (Figure 3). ETA displayed a significant portion of CNS demyelination (41%), followed by peripheral demyelination (22%), neuropathies (14.9%), myelopathy (11%), inflammatory demyelination (3.7%), non-demyelinating CNS inflammation (3.7%), and neurodegenerative diseases (3.7%). CZP was mainly associated with myelopathy (50%) and CNS demyelination (50%). GOL induced CND demyelination in 40% of cases and myelopathy (40%), followed by peripheral demyelination (20%). The neurological adverse effects after ADA treatment were mainly CNS (36%) and peripheral demyelination (15%), followed by LSS (10%) neuropathies (7.7%), meningitis (7.7%), myelopathy (7.7%), autoimmune encephalitis (5.1%), Henoch–Schönlein purpura (2.7%), inflammatory demyelination (2.7%), MRS (2.7%), and neurodegenerative diseases (2.7%) (Figure 3).
The TNF-α inhibitor discontinuation led, in the majority of cases, to partial or total resolution of the secondary diagnosed condition; only one case was reported where despite the adverse effects, adalimumab was continued due to its high efficiency in treating the primary condition, Crohn’s disease. INFL showed a balanced distribution of outcomes, with significant resolution as the predominant one (53%); the rest were as follows: 10% mild improvement, 15% no improvement, 2.5% not reported, partial resolution 18%, and significant improvement 2.5% (Figure 4). ADA was the only TNF-α inhibitor that led to the worsening of the neurological symptoms (2.6%) in parallel with a high frequency of significant resolution cases (59%). ETA displays a variety of outcomes with numerous instances of significant resolution (34.5%), significant improvement (24.2%), and mild improvement (22%) in the outcomes of the patients. CZP was only associated with partial (50%) and significant resolution (50%). GOL discontinuation resulted in significant (80%) and mild improvement (20%) of secondary disease (Figure 4).

5. Discussion

The administration of TNF-α inhibitors can be associated with various types of neurological damage, although the risk of such events is rather low. In numerous cases, TNF-α inhibitors triggered inflammatory reactions in the CNS with associated demyelinating or non-demyelinating conditions. However, there is much debate about whether this treatment is really the cause of such reactions since inflammatory events in the CNS have been reported even before the introduction of biological therapies in autoimmune diseases which by themselves may be responsible for CNS inflammatory processes. It is of particular importance to recognize the intrinsic neurological manifestations of the autoimmune condition itself in order to correctly establish a causality relationship between administered drugs and clinical symptoms; for example, inflammatory bowel diseases are mostly associated with cerebrovascular or demyelinating CNS disease, as well as peripheral neuropathy [150].
One study conducted in 2011 in 10 patients revealed the development of demyelination events as a result of TNF-α inhibitor therapy with a clear distinction from sporadic multiple sclerosis; most symptoms occurred within one year from the beginning of the therapy [151]. In 2020, Kunchock et al. evaluated a lot of 212 patients with autoimmune diseases divided in two groups according to the presence/absence of CNS inflammation; the study reported that the administration of TNF-α inhibitors was clearly associated with higher risk of both demyelinating or non-demyelinating events, in particular in patients diagnosed with rheumatoid arthritis, but could not conclude whether this association occurred separately or represented an exacerbated inflammation process [152]. White matter lesions in the brain parenchyma were identified through magnetic resonance imaging in a patient exhibiting neurological symptoms such as disorientation and confusion which were diagnosed as posterior reversible encephalopathy syndrome that was linked mainly to the existence of an autoimmune condition, rheumatoid arthritis, but also to the administration of immunosuppressive medication, in this case infliximab [153]. The loss of brain parenchyma integrity following TNF-α inhibitor therapy was confirmed through advanced MRI techniques, but neither inflammation nor demyelination could be identified as potential causes [154]; similarly, a fulminant and fatal CNS adverse event occurred after the first dose of infliximab in a pediatric patient who was diagnosed with ischemic and hemorrhagic central processes instead of demyelination or inflammation but was labeled as drug-related [155].
In our analysis, most adverse effects were reported for etanercept, infliximab, and adalimumab; their longer usage period in comparison to golimumab and certolizumab may be a contributing factor. Also, most neurological conditions were classified as demyelinating events, including clear diagnosis as multiple sclerosis. MS is a complex pathology that involves demyelination and axonal degeneration at the central level mainly due to immunological factors; the pathogenesis of the disease consists of an autoimmune attack against myelinated axons mediated by inflammatory cytokines and chemokines that facilitate the recruitment of infiltrating inflammatory cells, which are further activated in the CNS, where they react with immune and neuronal cells, thus triggering demyelination and axonal degeneration [156]. The mechanisms identified in the pathogenesis of MS are illustrated in Figure 5.
Demyelinating events may occur as a development of the autoimmune disease itself; in fact, the occurrence of a single acute neurological symptom, which is common in MS, may be an early sign of an autoimmune condition, thus making the differential diagnosis highly challenging [157]. In our analysis, the most frequently encountered primary diseases were rheumatoid arthritis, Crohn’s disease, and psoriasis. In an excellent review published in 2018, Atzeni et al. summarized the direct and indirect effects of rheumatoid arthritis on the CNS [158]. Briefly, the systemic inflammation in RA may directly be responsible for meningitis, vasculitis, and rheumatoid nodules, although such events are rather uncommon; indirectly, the prolonged systemic inflammation in RA may facilitate the development of CNS comorbidities, including vascular, neurodegenerative and demyelinating events. However, demyelinating conditions, including MS, rarely occur in RA patients who have not been treated with TNF-α inhibitors; also, the occurrence of extrapyramidal syndromes is low, but this could be attributed to anti-inflammatory drugs used as chronic treatment in RA. RA may also be accompanied by seizures, which display an inverse relation with the use of NSAIDs; dementia and other neuro-psychiatric pathologies also occur with relatively high frequency in comparison to the general population. Generally, various types of peripheral neuropathies may develop in autoimmune rheumatic diseases; in our analysis, eight cases of Guillain–Barré syndrome were identified while the diagnosis of peripheral neuropathy was established in more than 35% of all cases. In Crohn’s disease, about 3% of the patients experience neurological involvement consisting of peripheral nerve disease, myelopathy, myopathy, and cerebrovascular events; non-enhancing lesions of the white matter of ischemic, atherosclerotic, vasculitic, or demyelinating nature can be seen in asymptomatic IBD patients [159]. CD may co-exist with MS and a family/personal history of one of the two diseases may increase the risk of developing the other; this may be attributed to genes shared by several autoimmune pathologies which subsequently cluster in certain families and to contributing environmental factors. Regarding psoriasis, the pathogenesis of this autoimmune skin pathology shows multiple links with several neurodegenerative conditions, presumably due to the common germ layer, the ectoderm; additionally, the common background includes genetic factors, similar inflammatory mediators, oxidative stress, and various associations with metabolic disorders [160]. In ankylosing spondylitis which also occurs rather frequently as the primary disease in our study, the most common neurological symptoms are caused by spinal cord involvement due to impingement of bones and spinal stenosis [161]; however, subclinical neurological complications are more frequently seen compared to clinically manifested symptoms [162].
The common measure in all reviewed cases was the discontinuation of TNF-α inhibitor therapy with or without the administration of additional treatment, usually consisting of corticosteroids, human immunoglobulin, and plasmapheresis. The most prevalent outcome after the discontinuation of the TNF-α inhibitor was the resolution or partial resolution of all neurological symptoms within various periods, usually ranging from a few days to a few months; only 16.3% of all cases showed no/limited response with one case of worsening neurological symptoms. Such symptom progress supports the hypothesis that biological therapy is directly related to the occurrence of neurological events. However, in all three cases of ALS development, the TNF-α inhibitor discontinuation did not induce any symptom improvement, and the disease progress finally led to the patient’s death by respiratory insufficiency.
TNF-α is a cytokine with pleiotropic effects on various cells thus exerting a crucial role in triggering a normal immune response; its regulating activity makes it a key player in the pathogenesis of several inflammatory and autoimmune pathologies. It is a homotrimer protein containing 157 amino acids and generated by activated immune cells; in turn, TNF-α induces the production of other inflammatory mediators. TNF-α is synthesized as the transmembrane form which is processed into its soluble form in order to be released. Two types of receptors have been identified, TNFR1 and TNFR2; the first is present in all cells and tissues, being the key signaling receptor, while the second occurs in immune cells and performs limited functions. Although it binds to both types, the transmembrane TNF-α acts mainly through TNFR2, while its soluble form activates both types of receptors [163]. Its involvement in the pathogenesis of various autoimmune diseases encompasses multiple mechanisms, as depicted in Figure 6.
TNF-α inhibitors have been proposed as a treatment in MS based on the key role played by the TNF-α cytokine in MS pathogenesis; however, multiple studies have shown that not only did TNFα inhibitors fail to stop disease progress, but they even caused symptom exacerbation [164]. Recent studies, however, highlighted the difference between the two types of TNF receptors in terms of biological effects; while TNFR1 exerts pro-inflammatory and apoptotic effects, TNFR2 is associated with immunoregulatory, regenerative, and neuroprotective activities [165]. Therefore, a potential explanation for treatment failure in MS, as well as the adverse neurological events that occurred in other therapies, is the non-selective effect of TNF-α inhibitors currently in use, which counteracts the anti-inflammatory activity mediated by TNFR2 receptors. As support for this hypothesis stand the therapeutic effects exerted by atrosimab, a selective TNFR1 inhibitor, in neurodegenerative diseases in animal models [165]. Similarly, the preclinical evaluation of other TNFR1 inhibitors or DN-TNF muteins revealed superior outcomes to global TNF-α blockers in terms of less severe side effects or the potential to treat pathologies such as MS or other neurodegenerative conditions where non-selective TNFα inhibition is not recommended [166].
Other hypotheses regarding the controversial behaviors of TNF-α inhibitors were summarized in an excellent review conducted by Kaltsonoudis et al.; briefly, the following theories have been formulated: (1) TNF-α inhibitors increase the T-cell response by multiple mechanisms, followed by their entrance into the CNS and subsequent demyelination; (2) TNF-α inhibitors alter the downstream cytokine response by decreasing IL-10 and increasing IL-12 production, thus facilitating demyelination; (3) TNF-α inhibitors cause a peripheral reduction in TNF-α, while brain levels remain relatively unchanged due to the blood–brain barrier, which is difficult to cross; this may increase the level of TNFα in the brain and/or TNFR expression, thus inducing demyelinating lesions, and (4) TNF-α inhibitors may activate latent infections able to trigger autoimmune demyelinating events [167].
Controversially, we were able to identify a case where etanercept was successfully used for the treatment of ankylosing spondylitis combined with demyelinating myelitis; however, considering the already known risks of biological therapy, the administered dose was half (25 mg/week) of the routine dose (50 mg/week). The follow-up MRI scan (after 35 days) revealed a reduction in the vertebrae demyelinating lesion without the occurrence of new ones [168]; a further reduction in the dose to 25 mg every two weeks did not induce a relapse of either pathology. Another controversial case was reported, describing a patient who developed the Guillain–Barré syndrome after the initiation of infliximab therapy for psoriasis and ankylosing spondylitis [169]; the infliximab therapy was discontinued but then reinstated due to a severe relapse, however, at a lower dose that managed to control the autoimmune conditions. These reports suggest that the occurrence/severity of neurological events during TNF-α inhibitor therapy may be also related to the administered dose which was sufficient to control patient’s condition but low enough to avoid neurologic disease.

6. Conclusions and Future Perspectives

This study summarizes the neurological events reported during and after the administration of TNF-α inhibitors as treatment for various primary diseases. The profound involvement of TNF-α in the pathogenesis of MS suggested the potential of using TNF-α inhibitors as treatment for neurodegenerative diseases; far from producing beneficial effects, the drugs even exacerbated the existing symptoms. The debate regarding the true link between the occurrence of neurological events and the use of TNF-α inhibitors started almost immediately after the approval of the first such drug, etanercept, more than 20 years ago. During this time, both demyelinating and non-demyelinating events have been reported, with a large plethora of symptoms for all five approved TNF-α inhibitors. A number of theories were suggested in the attempt to explain the underlying mechanisms of such events; the most recent one is based on the existence of the two types of TNF-α receptors with opposite biological effects that triggered the discovery of selective TNFR1 inhibitors which induced superior outcomes to global TNF-α antagonists. The reported beneficial effects of TNF-α inhibitors in demyelinating diseases, although rare compared to the vast majority of neurological adverse effects, seem to be related to the administered dose, the authors concluding that it was high enough to control the primary disease and low enough to avoid triggering neurological events. Regardless, the general opinion in the field is that TNF-α inhibitors should be avoided in patients with a personal or familial history of demyelination (such as patients having first-degree relatives diagnosed with MS) due to their genetic proneness to develop such conditions even in the absence of an anti-TNF treatment; it is still possible that some patients have developed the disease anyway and the administration of the inhibitors only hastened it. Additionally, in countries where only TNF-α inhibitors are approved as advanced treatments in rheumatic diseases, clinicians should be aware of the potential neurological side effects and even the unmasking of a latent neurodegenerative condition with severe consequences; furthermore, a family history of autoimmune conditions may indicate increased susceptibility towards CNS demyelination, aspects that must be considered during the initial clinical evaluation of the patient. Constant monitoring is also necessary for patients undergoing TNF-α inhibitor therapy, in particular for those suffering from illnesses found to be correlated with more frequent side effects; such monitoring includes laboratory tests where dermatology patients exhibit less frequent abnormal results than gastroenterology or rheumatology patients, therefore requiring less frequent testing. Usually, laboratory evaluation should be performed at baseline and after 3, 6, and 12 months of treatment, followed by annual evaluation or more frequently depending on comorbidities, other medications, or medical experience (10.5144/0256-4947.2022.309). Full physical examination, brain MRI, and neurophysiology tests are essential both for the initial patient evaluation in order to identify preexisting demyelinating diseases and for the follow-up and monitoring of potential developing neurological complications. If any neurological symptoms occur, the patient should undergo a thorough neurological examination, including MRI scanning, that can confirm or exclude demyelination; if demyelination is confirmed, the TNF-α inhibitor should be discontinued and therapy adjusted to patient needs. Future studies will presumably lead to the discovery of other TNFR1 selective inhibitors with improved therapeutic outcomes, therefore reducing the risk of neurological adverse effects.

Author Contributions

Conceptualization, A.G. and C.B.; methodology, A.G. and C.S.; software, A.V. and M.V.; validation, A.B.; formal analysis, A.M.; data curation, A.V. and A.J.; writing—original draft preparation, A.G., A.B., A.M., and A.J.; writing—review and editing, M.V., C.S., and C.B.; visualization, A.G. and A.B.; supervision, C.B. All authors have read and agreed to the published version of the manuscript.

Funding

We would like to acknowledge the Victor Babes University of Medicine and Pharmacy Timisoara for their support in covering the costs of publication for this research paper.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

The original contributions presented in the study are included in the article, further inquiries can be directed to the corresponding author(s).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Coates, L.C.; Marzo-Ortega, H.; Bennett, A.N.; Emery, P. Anti-TNF therapy in ankylosing spondylitis: Insights for the clinician. Ther. Adv. Musculoskelet. Dis. 2010, 2, 37–43. [Google Scholar] [CrossRef] [PubMed]
  2. Pagnini, C.; Cominelli, F. Tumor Necrosis Factor’s Pathway in Crohn’s Disease: Potential for Intervention. Int. J. Mol. Sci. 2021, 22, 10273. [Google Scholar] [CrossRef] [PubMed]
  3. Horneff, G.; Minden, K.; Rolland, C.; Daly, A.C.H.; Borlenghi, C.; Ruperto, N. Efficacy and safety of TNF inhibitors in the treatment of juvenile idiopathic arthritis: A systematic literature review. Pediatr. Rheumatol. 2023, 21, 20. [Google Scholar] [CrossRef] [PubMed]
  4. Yost, J.; Gudjonsson, J.E. The role of TNF inhibitors in psoriasis therapy: New implications for associated comorbidities. F1000 Med. Rep. 2009, 1, 30. [Google Scholar] [CrossRef] [PubMed]
  5. Mantravadi, S.; Ogdie, A.; Kraft, W.K. Tumor necrosis factor inhibitors in psoriatic arthritis. Expert Rev. Clin. Pharmacol. 2017, 10, 899–910. [Google Scholar] [CrossRef]
  6. MA, X.; XU, S. TNF inhibitor therapy for rheumatoid arthritis. Biomed. Rep. 2013, 1, 177–184. [Google Scholar] [CrossRef]
  7. Aslam, N.; Lo, S.W.; Sikafi, R.; Barnes, T.; Segal, J.; Smith, P.J.; Limdi, J.K. A review of the therapeutic management of ulcerative colitis. Therap. Adv. Gastroenterol. 2022, 15, 175628482211381. [Google Scholar] [CrossRef]
  8. Leal, I.; Rodrigues, F.B.; Sousa, D.C.; Duarte, G.S.; Romão, V.C.; Marques-Neves, C.; Costa, J.; Fonseca, J.E. Anti-TNF Drugs for Chronic Uveitis in Adults—A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Front. Med. 2019, 6, 104. [Google Scholar] [CrossRef]
  9. dos Santos Nascimento, I.J.; da Silva-Júnior, E.F. TNF-α Inhibitors from Natural Compounds: An Overview, CADD Approaches, and their Exploration for Anti-inflammatory Agents. Comb. Chem. High Throughput Screen. 2022, 25, 2317–2340. [Google Scholar] [CrossRef]
  10. Majumder, S.; Rama Chaitanya Sreedhara, S.; Banerjee, S.; Chatterjee, S. TNF α Signaling Beholds Thalidomide Saga: A Review of Mechanistic Role of TNF-α Signaling Under Thalidomide. Curr. Top. Med. Chem. 2012, 12, 1456–1467. [Google Scholar] [CrossRef]
  11. Kotla, V.; Goel, S.; Nischal, S.; Heuck, C.; Vivek, K.; Das, B.; Verma, A. Mechanism of action of lenalidomide in hematological malignancies. J. Hematol. Oncol. 2009, 2, 36. [Google Scholar] [CrossRef]
  12. Zhengxiang, L.; Wang, B.; Liu, H.; Shi, L. TNF Inhibitor Pomalidomide Sensitizes Glioblastoma Cells to EGFR Inhibition. Ann. Clin. Lab. Sci. 2020, 50, 474–480. [Google Scholar]
  13. Singh, N.; Shreshtha, A.K.; Thakur, M.S.; Patra, S. Xanthine scaffold: Scope and potential in drug development. Heliyon 2018, 4, e00829. [Google Scholar] [CrossRef] [PubMed]
  14. Brustolim, D.; Ribeiro-dos-Santos, R.; Kast, R.E.; Altschuler, E.L.; Soares, M.B.P. A new chapter opens in anti-inflammatory treatments:The antidepressant bupropion lowers production of tumor necrosis factor-alpha and interferon-gamma in mice. Int. Immunopharmacol. 2006, 6, 903–907. [Google Scholar] [CrossRef] [PubMed]
  15. Johansen, C.B.; Jimenez-Solem, E.; Haerskjold, A.; Sand, F.L.; Thomsen, S.F. The Use and Safety of TNF Inhibitors during Pregnancy in Women with Psoriasis: A Review. Int. J. Mol. Sci. 2018, 19, 1349. [Google Scholar] [CrossRef]
  16. Patel, S.; Wadhwa, M. Therapeutic use of specific tumour necrosis factor inhibitors in inflammatory diseases including COVID-19. Biomed. Pharmacother. 2021, 140, 111785. [Google Scholar] [CrossRef]
  17. Pauline, O.; Robert, M.; Bernardeau, C.; Hlavaty, A.; Fusaroli, M.; Roustit, M.; Cracowski, J.-L.; Khouri, C. Assessment of Reported Adverse Events After Interchanging Between TNF-α Inhibitor Biosimilars in the WHO Pharmacovigilance Database. BioDrugs 2023, 37, 699–707. [Google Scholar] [CrossRef]
  18. Li, J.; Zhang, Z.; Wu, X.; Zhou, J.; Meng, D.; Zhu, P. Risk of Adverse Events After Anti-TNF Treatment for Inflammatory Rheumatological Disease. A Meta-Analysis. Front. Pharmacol. 2021, 12, 746396. [Google Scholar] [CrossRef]
  19. Muller, M.; D’Amico, F.; Bonovas, S.; Danese, S.; Peyrin-Biroulet, L. TNF Inhibitors and Risk of Malignancy in Patients with Inflammatory Bowel Diseases: A Systematic Review. J. Crohn’s Colitis 2021, 15, 840–859. [Google Scholar] [CrossRef]
  20. Yen, C.-Y.; Yu, S.-J.; Chen, Y.-M.; Lai, K.-L.; Wu, Y.-D.; Liao, E.-C.; Hsieh, C.-L. Mechanisms of Tumor Necrosis Factor-Alpha Inhibitor-Induced Systemic Lupus Erythematosus. Front. Med. 2022, 9, 870724. [Google Scholar] [CrossRef]
  21. Drosos, A.A.; Pelechas, E.; Kaltsonoudis, E.; Markatseli, T.E.; Voulgari, P.V. Biologic Therapies and Autoimmune Phenomena. Mediterr. J. Rheumatol. 2021, 32, 96. [Google Scholar] [CrossRef]
  22. U.S. Food and Drug Administration. Enbrel® (Etanercept) Solution for Subcutaneous Use; U.S. Food and Drug Administration: Silver Spring, MD, USA, 1998.
  23. Catrina, A.I.; Lampa, J.; Ernestam, S.; af Klint, E.; Bratt, J.; Klareskog, L.; Ulfgren, A.-K. Anti-tumour necrosis factor (TNF)-alpha therapy (etanercept) down-regulates serum matrix metalloproteinase (MMP)-3 and MMP-1 in rheumatoid arthritis. Rheumatology 2002, 41, 484–489. [Google Scholar] [CrossRef]
  24. Klimiuk, P.A.; Sierakowski, S.; Domyslawska, I.; Chwiecko, J. Effect of etanercept on serum levels of soluble cell adhesion molecules (sICAM-1, sVCAM-1, and sE-selectin) and vascular endothelial growth factor in patients with rheumatoid arthritis. Scand. J. Rheumatol. 2009, 38, 439–444. [Google Scholar] [CrossRef]
  25. Haraoui, B.; Bykerk, V. Etanercept in the treatment of rheumatoid arthritis. Ther. Clin. Risk Manag. 2007, 3, 99–105. [Google Scholar] [CrossRef]
  26. Rios Rodriguez, V.; Poddubnyy, D. Etanercept for the treatment of non-radiographic axial spondyloarthritis. Expert Rev. Clin. Immunol. 2016, 12, 493–500. [Google Scholar] [CrossRef]
  27. U.S. Food and Drug Administration. Infliximab for Injection, for Intravenous Use; U.S. Food and Drug Administration: Silver Spring, MD, USA, 1998.
  28. Elliott, M.J.; Maini, R.N.; Feldmann, M.; Kalden, J.R.; Antoni, C.; Smolen, J.S.; Leeb, B.; Breedveld, F.C.; Macfarlane, J.D.; Bijl, H. Randomised double-blind comparison of chimeric monoclonal antibody to tumour necrosis factor alpha (cA2) versus placebo in rheumatoid arthritis. Lancet 1994, 344, 1105–1110. [Google Scholar] [CrossRef]
  29. Taylor, P.C.; Peters, A.M.; Paleolog, E.; Chapman, P.T.; Elliott, M.J.; McCloskey, R.; Feldmann, M.; Maini, R.N. Reduction of chemokine levels and leukocyte traffic to joints by tumor necrosis factor alpha blockade in patients with rheumatoid arthritis. Arthritis Rheum. 2000, 43, 38–47. [Google Scholar] [CrossRef]
  30. Paleolog, E.M.; Hunt, M.; Elliott, M.J.; Feldmann, M.; Maini, R.N.; Woody, J.N. Deactivation of vascular endothelium by monoclonal anti-tumor necrosis factor alpha antibody in rheumatoid arthritis. Arthritis Rheum. 1996, 39, 1082–1091. [Google Scholar] [CrossRef]
  31. Soliman, E.; El-tantawi, G.; Matrawy, K.; Aldawoudy, A.; Naguib, A. Local infliximab injection of sacroiliac joints in non-radiographic axial spondyloarthritis: Impact on clinical and magnetic resonance imaging parameters of disease activity. Mod. Rheumatol. 2015, 25, 421–426. [Google Scholar] [CrossRef]
  32. U.S. Food and Drug Administration. HUMIRA® (Adalimumab) Injection, for Subcutaneous Use; U.S. Food and Drug Administration: Silver Spring, MD, USA, 2002.
  33. Zdanowska, N.; Owczarczyk-Saczonek, A.; Czerwińska, J.; Nowakowski, J.J.; Kozera-Żywczyk, A.; Owczarek, W.; Zdanowski, W.; Placek, W. Methotrexate and Adalimumab Decrease the Serum Levels of Cardiovascular Disease Biomarkers (VCAM-1 and E-Selectin) in Plaque Psoriasis. Medicina 2020, 56, 473. [Google Scholar] [CrossRef]
  34. Goto, D.; Sumida, T. Adalimumab. Nihon Rinsho 2002, 60, 2384–2389. [Google Scholar]
  35. Shen, C.; Assche, G.V.; Colpaert, S.; Maerten, P.; Geboes, K.; Rutgeerts, P.; Ceuppens, J.L. Adalimumab induces apoptosis of human monocytes: A comparative study with infliximab and etanercept. Aliment. Pharmacol. Ther. 2005, 21, 251–258. [Google Scholar] [CrossRef]
  36. Oberoi, R.; Schuett, J.; Schuett, H.; Koch, A.-K.; Luchtefeld, M.; Grote, K.; Schieffer, B. Targeting Tumor Necrosis Factor-α with Adalimumab: Effects on Endothelial Activation and Monocyte Adhesion. PLoS ONE 2016, 11, e0160145. [Google Scholar] [CrossRef]
  37. Timis, T.-L.; Beni, L.; Florian, I.-A.; Orăsan, M.; Orăsan, R.I. Prevalence of metabolic syndrome and chronic inflammation in psoriasis before and after biologic therapy: A prospective study. Med. Pharm. Rep. 2023, 96, 368–383. [Google Scholar] [CrossRef]
  38. Rojas-Carabali, W.; Boada-Robayo, L.; Chacón-Zambrano, D.; Criollo Porras, E.; Kerguelén Dumar, V.; De-la-Torre, A. Multiple Sclerosis in a Patient with Intermediate Uveitis and Juvenile Idiopathic Arthritis Treated with Adalimumab: A Case Report. Ocul. Immunol. Inflamm. 2023, 31, 1873–1876. [Google Scholar] [CrossRef]
  39. Alnasser Alsukhni, R.; Jriekh, Z.; Aboras, Y. Adalimumab Induced or Provoked MS in Patient with Autoimmune Uveitis: A Case Report and Review of the Literature. Case Rep. Med. 2016, 2016, 1423131. [Google Scholar] [CrossRef]
  40. Burness, C.B.; Deeks, E.D. Adalimumab: In non-radiographic axial spondyloarthritis. Drugs 2012, 72, 2385–2395. [Google Scholar] [CrossRef]
  41. U.S. Food and Drug Administration. SIMPONI (Gollmumab) Injection, Solution for Subcutaneous Use; U.S. Food and Drug Administration: Silver Spring, MD, USA, 2009.
  42. Kanbe, K.; Chiba, J.; Inoue, Y.; Taguchi, M.; Yabuki, A. Predictive factors related to the efficacy of golimumab in patients with rheumatoid arthritis. Clin. Med. Insights. Arthritis Musculoskelet. Disord. 2015, 8, 25–32. [Google Scholar] [CrossRef]
  43. Doyle, M.K.; Rahman, M.U.; Frederick, B.; Birbara, C.A.; de Vries, D.; Toedter, G.; Wu, X.; Chen, D.; Ranganath, V.K.; Westerman, M.E.; et al. Effects of subcutaneous and intravenous golimumab on inflammatory biomarkers in patients with rheumatoid arthritis: Results of a phase 1, randomized, open-label trial. Rheumatology 2013, 52, 1214–1219. [Google Scholar] [CrossRef]
  44. Toussirot, E.; Vauchy, C.; Binda, D.; Michel, F. Golimumab in radiographic and nonradiographic axial spondyloarthritis: A review of clinical trials. Drug Des. Devel. Ther. 2016, 10, 2087–2094. [Google Scholar] [CrossRef]
  45. U.S. Food and Drug Administration. CIMZIA (Certolizumab Pegol) for Injection, for Subcutaneous Use; U.S. Food and Drug Administration: Silver Spring, MD, USA, 2008.
  46. Patel, V.K.; Ghosh, S. Certolizumab pegol in Crohn’s disease. Drugs Today 2008, 44, 837–844. [Google Scholar] [CrossRef]
  47. Robinson, P.C.; Maksymowych, W.P.; Gensler, L.S.; Hall, S.; Rudwaleit, M.; Hoepken, B.; Bauer, L.; Kumke, T.; Kim, M.; de Peyrecave, N.; et al. Certolizumab Pegol Efficacy in Patients With Non-Radiographic Axial Spondyloarthritis Stratified by Baseline MRI and C-Reactive Protein Status: An Analysis From the C-axSpAnd Study. ACR Open Rheumatol. 2022, 4, 794–801. [Google Scholar] [CrossRef]
  48. Sedger, L.M.; McDermott, M.F. TNF and TNF-receptors: From mediators of cell death and inflammation to therapeutic giants—Past, present and future. Cytokine Growth Factor Rev. 2014, 25, 453–472. [Google Scholar] [CrossRef]
  49. Carpentier, I.; Coornaert, B.; Beyaert, R. Function and Regulation of Tumor Necrosis Factor Receptor Type 2. Curr. Med. Chem. 2004, 11, 2205–2212. [Google Scholar] [CrossRef]
  50. Kist, M.; Kőműves, L.G.; Goncharov, T.; Dugger, D.L.; Yu, C.; Roose-Girma, M.; Newton, K.; Webster, J.D.; Vucic, D. Impaired RIPK1 ubiquitination sensitizes mice to TNF toxicity and inflammatory cell death. Cell Death Differ. 2021, 28, 985–1000. [Google Scholar] [CrossRef]
  51. Brenner, D.; Blaser, H.; Mak, T.W. Regulation of tumour necrosis factor signalling: Live or let die. Nat. Rev. Immunol. 2015, 15, 362–374. [Google Scholar] [CrossRef]
  52. Aggarwal, B.B.; Gupta, S.C.; Kim, J.H. Historical perspectives on tumor necrosis factor and its superfamily: 25 years later, a golden journey. Blood 2012, 119, 651–665. [Google Scholar] [CrossRef]
  53. Vanden Berghe, T.; Linkermann, A.; Jouan-Lanhouet, S.; Walczak, H.; Vandenabeele, P. Regulated necrosis: The expanding network of non-apoptotic cell death pathways. Nat. Rev. Mol. Cell Biol. 2014, 15, 135–147. [Google Scholar] [CrossRef]
  54. Wajant, H.; Siegmund, D. TNFR1 and TNFR2 in the Control of the Life and Death Balance of Macrophages. Front. Cell Dev. Biol. 2019, 7, 91. [Google Scholar] [CrossRef]
  55. Lin, Y.; Bai, L.; Chen, W.; Xu, S. The NF-kappaB activation pathways, emerging molecular targets for cancer prevention and therapy. Expert Opin. Ther. Targets 2010, 14, 45–55. [Google Scholar] [CrossRef]
  56. Pflug, K.M.; Sitcheran, R. Targeting NF-κB-Inducing Kinase (NIK) in Immunity, Inflammation, and Cancer. Int. J. Mol. Sci. 2020, 21, 8470. [Google Scholar] [CrossRef]
  57. Li, M.; Zhang, X.; Bai, X.; Liang, T. Targeting TNFR2: A Novel Breakthrough in the Treatment of Cancer. Front. Oncol. 2022, 12, 862154. [Google Scholar] [CrossRef]
  58. Cabal-Hierro, L.; Rodríguez, M.; Artime, N.; Iglesias, J.; Ugarte, L.; Prado, M.A.; Lazo, P.S. TRAF-mediated modulation of NF-kB AND JNK Activation by TNFR2. Cell. Signal. 2014, 26, 2658–2666. [Google Scholar] [CrossRef]
  59. Dziadzio, M.; Reddy, V.; Rahman, S.; Mummery, C.; Keat, A. Is TNFα really a good therapeutic target in motoneuronal degeneration? A case of amyotrophic lateral sclerosis in a patient with RA receiving infliximab. Rheumatology 2006, 45, 1445–1446. [Google Scholar] [CrossRef]
  60. Loustau, V.; Foltz, V.; Poulain, C.; Rozenberg, S.; Bruneteau, G. Diagnosis of amyotrophic lateral sclerosis in a patient treated with TNFα blockers for ankylosing spondylitis: Fortuitus association or new side effect of TNFα blockers? Jt. Bone Spine 2009, 76, 213–214. [Google Scholar] [CrossRef]
  61. Padovan, M.; Caniatti, L.M.; Trotta, F.; Govoni, M. Concomitant rheumatoid arthritis and amyotrophic lateral sclerosis: Report of two new cases and review of literature. Rheumatol. Int. 2011, 31, 715–719. [Google Scholar] [CrossRef]
  62. Fernández Alvarez, P.; Maldonado Pérez, B.; Castro Laria, L.; Argüelles-Arias, F. Autoimmune Encephalitis During Treatment With Adalimumab: A Case Report in Crohn’s Disease. Inflamm. Bowel Dis. 2021, 27, e40–e41. [Google Scholar] [CrossRef]
  63. Kim, M.; Kim, M.-J.; Suh, Y.S.; Kim, H.-O.; Lee, S.-I.; Cheon, Y.-H. Bickerstaff brainstem encephalitis in a patient with ankylosing spondylitis on tumour necrosis factor-alpha inhibitor. Clin. Exp. Rheumatol. 2018, 36, 343. [Google Scholar]
  64. Condamina, M.; Diaz, E.; Jamart, C.; Loget, J.; Durlach, A.; Salmon, J.-H.; Cadiot, G.; Viguier, M. Severe Attack of Henoch-Schönlein Purpura With Neurological Involvement During Adalimumab Treatment for Crohn’s Disease. J. Crohn’s Colitis 2020, 14, 538–542. [Google Scholar] [CrossRef]
  65. Faivre, A.; Franques, J.; De Paula, A.M.; Gutierrez, M.; Bret, S.; Aubert, S.; Attarian, S.; Pouget, J. Acute motor and sensory axonal neuropathy and concomitant encephalopathy during tumor necrosis factor-alpha antagonist therapy. J. Neurol. Sci. 2010, 291, 103–106. [Google Scholar] [CrossRef]
  66. Oh, S.J.; Kwon, Y.N.; Lee, C.K.; Lee, J.S. Anti-NMDAR encephalitis in Crohn’s disease undergoing long-term infliximab treatment: A case report. Front. Immunol. 2022, 13, 957575. [Google Scholar] [CrossRef]
  67. Kumar, D.; Bouldin, T.W.; Berger, R.G. A case of progressive multifocal leukoencephalopathy in a patient treated with infliximab. Arthritis Rheum. 2010, 62, 3191–3195. [Google Scholar] [CrossRef]
  68. Oumerzouk, J.; Ahizoune, A.; Raggabi, A.; Mnaili, A.; Abida, N.; Benkirane, A.; Semlali, A.; Bourazza, A. Encéphalomyélite aiguë disséminée associée à une maladie de Crohn sous infliximab. Presse Med. 2017, 46, 975–978. [Google Scholar] [CrossRef]
  69. Ray, M.; Curtis, J.R.; Baddley, J.W. A case report of progressive multifocal leucoencephalopathy (PML) associated with adalimumab. Ann. Rheum. Dis. 2014, 73, 1429–1430. [Google Scholar] [CrossRef]
  70. Çimen Güneş, E.; Çolak, S.; Tekgöz, E.; Çınar, M.; Yılmaz, S. Golimumab-induced posterior reversible encephalopathy syndrome (PRES): A case-based review. Clin. Rheumatol. 2023, 42, 3407–3410. [Google Scholar] [CrossRef]
  71. Yamamoto, M.; Takahashi, H.; Wakasugi, H.; Sukawa, Y.; Saito, M.; Suzuki, C.; Naishiro, Y.; Yamamoto, H.; Shinomura, Y.; Imai, K. Leukoencephalopathy during administration of etanercept for refractory rheumatoid arthritis. Mod. Rheumatol. 2007, 17, 72–74. [Google Scholar] [CrossRef]
  72. Yıldırım, R.; Cansu, D.Ü.; Arık, D.; Saylısoy, S.; Korkmaz, C. Aseptic meningitis in rheumatoid arthritis after anti-TNF administration: A case-based literature review. Rheumatol. Int. 2021, 41, 1845–1853. [Google Scholar] [CrossRef]
  73. Ahmed, M.; Luggen, M.; Herman, J.H.; Weiss, K.L.; Decourten-Myers, G.; Quinlan, J.G.; Khanna, D. Hypertrophic pachymeningitis in rheumatoid arthritis after adalimumab administration. J. Rheumatol. 2006, 33, 2344–2346. [Google Scholar]
  74. Akça, Ü.K.; Sökmen, O.; Bölek, E.Ç.; Demir, S.; Kılıç, L.; Çevik, I.Ü.; Bilginer, Y. A case report of intracranial hypertension and aseptic meningitis: Anti-tumor necrosis factor associated or juvenile idiopathic arthritis related. Turk. J. Pediatr. 2021, 63, 522–526. [Google Scholar] [CrossRef]
  75. Barreras, P.; Mealy, M.A.; Pardo, C.A. TNF-alpha inhibitor associated myelopathies: A neurological complication in patients with rheumatologic disorders. J. Neurol. Sci. 2017, 373, 303–306. [Google Scholar] [CrossRef]
  76. Caracseghi, F.; Izquierdo-Blasco, J.; Sanchez-Montanez, A.; Melendo-Perez, S.; Roig-Quilis, M.; Modesto, C. Etanercept-Induced Myelopathy in a Pediatric Case of Blau Syndrome. Case Rep. Rheumatol. 2011, 2011, 134106. [Google Scholar] [CrossRef]
  77. Kudsi, M.; Shahada, Z.; Haidar, G.; Safiah, M.H.; Khalayli, N. Golimumab therapy-induced isolated myelitis in a Behcet’s disease patient: A case report. Ann. Med. Surg. 2023, 85, 951–954. [Google Scholar] [CrossRef]
  78. Kon, T.; Hasui, K.; Suzuki, C.; Nishijima, H.; Tomiyama, M. Isolated myelitis in a patient with Behcet’s disease during golimumab therapy. J. Neuroimmunol. 2021, 354, 577533. [Google Scholar] [CrossRef]
  79. Vadikolias, K.; Kouklakis, G.; Heliopoulos, I.; Argyropoulou, P.; Papanas, N.; Tzilonidou, M.; Prassopoulos, P.; Piperidou, H. Acute paraplegia after the initiation of anti-tumour necrosis factor-α therapy for Crohn’s disease. Eur. J. Gastroenterol. Hepatol. 2007, 19, 159–162. [Google Scholar] [CrossRef]
  80. Hou, C.; Azzi, E.; Salmon, A.; Osmont, M.-N.; Perdriger, A. Reversible anti-TNFα treatment induced dementia: A case report. Jt. Bone Spine 2021, 88, 105044. [Google Scholar] [CrossRef]
  81. Bellesi, M.; Logullo, F.; Bella, P.; Provinciali, L. CNS demyelination during anti–tumor necrosis factor alpha therapy. J. Neurol. 2006, 253, 668–669. [Google Scholar] [CrossRef]
  82. Berthelot, C.N.; George, S.J.; Hsu, S. Distal lower extremity paresthesia and foot drop developing during adalimumab therapy. J. Am. Acad. Dermatol. 2005, 53, S260–S262. [Google Scholar] [CrossRef]
  83. Boggs, J.M.E.; Barnes, L. Demyelination during anti-tumour necrosis factor therapy for psoriasis. Clin. Exp. Dermatol. 2018, 43, 577–578. [Google Scholar] [CrossRef]
  84. Hamon, M.A.; Nicolas, G.; Deviere, F.; Letournel, F.; Dubas, F. Demyelinating neuropathy during anti-TNF alpha treatment with a review of the literature. Rev. Neurol. 2007, 163, 1232–1235. [Google Scholar] [CrossRef]
  85. Hare, N.C.; Hunt, D.P.J.; Venugopal, K.; Ho, G.-T.; Beez, T.; Lees, C.W.; Gibson, R.; Weller, B.; Satsangi, J. Multiple sclerosis in the context of TNF blockade and inflammatory bowel disease. QJM 2014, 107, 51–55. [Google Scholar] [CrossRef]
  86. Onasanya, J.; Beach, R.A. A case report about adalimumab-induced radiologically isolated syndrome: A significant side effect of tumour necrosis factor inhibitors. SAGE Open Med. Case Rep. 2021, 9, 2050313X2110030. [Google Scholar] [CrossRef]
  87. Sršen, S. How to Treat Patients after Serious Adverse Effects Caused by TNF Inhibitors? Acta Clin. Croat. 2020, 59, 161–165. [Google Scholar] [CrossRef]
  88. Barešić, M.; Reihl Crnogaj, M.; Zadro, I.; Anić, B. Demyelinating disease (multiple sclerosis) in a patient with psoriatic arthritis treated with adalimumab: A case-based review. Rheumatol. Int. 2021, 41, 2233–2239. [Google Scholar] [CrossRef]
  89. Mercieca, C.; Vella, N.; Borg, A.A. Demyelination during anti-TNFα therapy for ankylosing spondylitis. Mod. Rheumatol. 2012, 22, 303–307. [Google Scholar] [CrossRef]
  90. Luo, W.; Li, R.; Chang, Y.; She, H.; Kermode, A.G.; Qiu, W. Myelin oligodendrocyte glycoprotein antibody-associated disorders coexisting with ankylosing spondylitis: Potential association between demyelination and tumor necrosis factor inhibitors. Mult. Scler. Relat. Disord. 2021, 51, 102889. [Google Scholar] [CrossRef]
  91. Cereda, C.; Zecca, C.; Mazzucchelli, L.; Valci, L.; Staedler, C.; Bassetti, C.; Gobbi, C. Tumefactive demyelinating lesions during etanercept treatment requiring decompressive hemicraniectomy. Mult. Scler. J. 2013, 19, 820–823. [Google Scholar] [CrossRef]
  92. Cay, H.F.; Gungor, H.A.; Sezer, I.; Kacar, C.; Balci, N. Adverse effect of TNF-alpha blocker? Demyelination in an ankylosing spondylitis patient: A case report. J. Clin. Pharm. Ther. 2006, 31, 645–648. [Google Scholar] [CrossRef]
  93. Sicotte, N.L.; Voskuhl, R.R. Onset of multiple sclerosis associated with anti-TNF therapy. Neurology 2001, 57, 1885–1888. [Google Scholar] [CrossRef]
  94. Winkelmann, A.; Patejdl, R.; Wagner, S.; Benecke, R.; Zettl, U.K. Cerebral MRI lesions and anti-tumor necrosis factor-alpha therapy. J. Neurol. 2008, 255, 109–114. [Google Scholar] [CrossRef]
  95. Katsanos, A.H.; Kosmidou, M.; Giannopoulos, S. Vertebral artery hypoplasia in posterior circulation cerebral ischemia. Clin. Neurol. Neurosurg. 2013, 115, 1194–1195. [Google Scholar] [CrossRef]
  96. Yetkin, M.F.; Kendirci, M.; İsmailoğullari, S.; Gültekin, M.; Baydemir, R.; Sarilar, A.Ç.; Köse, E.G.; Akcakoyunlu, M.; Erbay, Ü.Z.; Erol, K.; et al. A case of fulminant demyelinating disease associated with golimumab treatment. Clin. Neurol. Neurosurg. 2020, 195, 105887. [Google Scholar] [CrossRef]
  97. Ruiz-Jimeno, T.; Carvajal, A.; Mata, C.; Aurrecoechea, E. Demyelinating disease in a patient with psoriatic arthritis and family history of multiple sclerosis treated with infliximab. J. Rheumatol. 2006, 33, 1457–1458. [Google Scholar]
  98. Kristensen, L.; Lambertsen, K.; Nguyen, N.; Byg, K.-E.; Nielsen, H. The Role of Non-Selective TNF Inhibitors in Demyelinating Events. Brain Sci. 2021, 11, 38. [Google Scholar] [CrossRef]
  99. Dubcenco, E.; Ottaway, C.A.; Chen, D.L.; Baker, J.P. Neurological symptoms suggestive of demyelination in Crohn’s disease after infliximab therapy. Eur. J. Gastroenterol. Hepatol. 2006, 18, 565–566. [Google Scholar] [CrossRef]
  100. Freeman, H.J.; Flak, B. Demyelination-like syndrome in Crohn’s disease after infliximab therapy. Can. J. Gastroenterol. 2005, 19, 313–316. [Google Scholar] [CrossRef]
  101. Papadia, M.; Herbort, C.P. Infliximab-induced Demyelination Causes Visual Disturbance Mistaken for Recurrence of HLA-B27-related Uveitis. Ocul. Immunol. Inflamm. 2010, 18, 482–484. [Google Scholar] [CrossRef]
  102. Tanno, M.; Nakamura, I.; Kobayashi, S.; Kurihara, K.; Ito, K. New-onset demyelination induced by infliximab therapy in two rheumatoid arthritis patients. Clin. Rheumatol. 2006, 25, 929–933. [Google Scholar] [CrossRef]
  103. Thomas, C.W.J.; Weinshenker, B.G.; Sandborn, W.J. Demyelination during anti-tumor necrosis factor alpha therapy with infliximab for Crohn’s disease. Inflamm. Bowel Dis. 2004, 10, 28–31. [Google Scholar] [CrossRef]
  104. Honda, Y.; Otsuka, A.; Egawa, G.; Inoue, Y.; Kuzuya, A.; Takahashi, R.; Miyachi, Y.; Kabashima, K. Multiple neurological abnormalities, including pontine hemorrhage, multiple sclerosis and aseptic meningitis, during anti-TNF-α therapy in psoriatic arthritis. Eur. J. Dermatol. 2015, 25, 487–488. [Google Scholar] [CrossRef]
  105. Matsumoto, T.; Nakamura, I.; Miura, A.; Momoyama, G.; Ito, K. New-onset multiple sclerosis associated with adalimumab treatment in rheumatoid arthritis: A case report and literature review. Clin. Rheumatol. 2013, 32, 271–275. [Google Scholar] [CrossRef]
  106. Uygunoğlu, U.; Uluduz, D.; Taşçılar, K.; Saip, S. Multiple sclerosis during adalimumab treatment in a case with ankylosing spondylitis. Rheumatol. Int. 2014, 34, 141–143. [Google Scholar] [CrossRef]
  107. Mir Subías, A.; García-López, S.; Sebastián Torres, B.; Ollero Domenche, L.; García Gámez, A.; Gomollón, F. Esclerosis múltiple como efecto adverso de los agentes antifactor de necrosis tumoral alfa: Una complicación infrecuente pero relevante de infliximab en la enfermedad de Crohn. Gastroenterol. Hepatol. 2013, 36, 81–85. [Google Scholar] [CrossRef] [PubMed]
  108. Robinson, M.R.; Korman, N.J. Psoriasis and Co-Morbidities: Approach to Treatment with Biologics. Psoriasis Forum 2006, 12, 10–15. [Google Scholar] [CrossRef]
  109. Theibich, A.; Dreyer, L.; Magyari, M.; Locht, H. Demyelinizing neurological disease after treatment with tumor necrosis factor alpha-inhibiting agents in a rheumatological outpatient clinic: Description of six cases. Clin. Rheumatol. 2014, 33, 719–723. [Google Scholar] [CrossRef]
  110. Napolitano, M.; Balato, N.; Ayala, F.; Cirillo, T.; Balato, A. Multiple sclerosis following anti-tumor necrosis factor-alpha therapy for psoriasis: First case in Italy? Ital. J. Dermatol. Venereol. 2018, 153, 567–572. [Google Scholar] [CrossRef]
  111. da Silva, L.G.M.; Sasso, L.S.; Nelli, C.J.; Bernardes Filho, F.; de Abreu, M.A.M.M. Optic neuritis due to immunobiologics: First Brazilian case report. An. Bras. Dermatol. 2013, 88, 162–165. [Google Scholar] [CrossRef]
  112. Aungsumart, S.; Apiwattanakul, M. Concurrent anterior and posterior visual pathway impairment in a patient treated with infliximab. J. Neurol. Sci. 2016, 367, 22–23. [Google Scholar] [CrossRef]
  113. Ban, B.H.; Crowe, J.L. Oculomotor Nerve Demyelination Secondary to Certolizumab Pegol. JCR J. Clin. Rheumatol. 2018, 24, 234–236. [Google Scholar] [CrossRef]
  114. Yokoyama, W.; Takada, K.; Miyasaka, N.; Kohsaka, H. Myelitis and optic neuritis induced by a long course of etanercept in a patient with rheumatoid arthritis. BMJ Case Rep. 2014, 2014, bcr2014205779. [Google Scholar] [CrossRef]
  115. Drury, J.; Hickman, S.J. Internuclear Ophthalmoplegia Associated with Anti-TNFα Medication. Strabismus 2015, 23, 30–32. [Google Scholar] [CrossRef] [PubMed]
  116. Faillace, C.; de Almeida, J.R.M.; de Carvalho, J.F. Optic neuritis after infliximab therapy. Rheumatol. Int. 2013, 33, 1101–1103. [Google Scholar] [CrossRef] [PubMed]
  117. Durel, C.-A.; Feurer, E.; Pialat, J.-B.; Berthoux, E.; Chapurlat, R.D.; Confavreux, C.B. Etanercept may induce neurosarcoidosis in a patient treated for rheumatoid arthritis. BMC Neurol. 2013, 13, 212. [Google Scholar] [CrossRef]
  118. Berrios, I.; Jun-O’Connell, A.; Ghiran, S.; Ionete, C. A case of neurosarcoidosis secondary to treatment of etanercept and review of the literature. BMJ Case Rep. 2015, 2015, bcr2014208188. [Google Scholar] [CrossRef] [PubMed]
  119. Jarand, J.; Zochodne, D.W.; Martin, L.O.; Voll, C. Neurological complications of infliximab. J. Rheumatol. 2006, 33, 1018–1020. [Google Scholar] [PubMed]
  120. Alexopoulou, A.; Koskinas, J.; Soultati, A.; Katsaounis, P.; Kilidireas, K.; Papageorgiou, C.; Antoniou, C.; Katsambas, A.; Archimandritis, A. Acute bilateral phrenic neuropathy following treatment with adalimumab. Clin. Rheumatol. 2009, 28, 1337–1340. [Google Scholar] [CrossRef] [PubMed]
  121. Alshekhlee, A.; Basiri, K.; Miles, J.D.; Ahmad, S.A.; Katirji, B. Chronic inflammatory demyelinating polyneuropathy associated with tumor necrosis factor-α antagonists. Muscle Nerve 2010, 41, 723–727. [Google Scholar] [CrossRef]
  122. Ahmed, Z.; Powell, R.; Llewelyn, G.; Anstey, A. Chronic inflammatory demyelinating polyradiculoneuropathy complicating anti TNF therapy for chronic plaque psoriasis. Case Rep. 2011, 2011, bcr0820114674. [Google Scholar] [CrossRef]
  123. McGinty, R.N.; McNamara, B.; Moore, H. DADS neuropathy associated with anti-TNF-α therapy. BMJ Case Rep. 2015, 2015, bcr2015211781. [Google Scholar] [CrossRef]
  124. de Frutos-Lezaun, M.; Bidaguren, A.; de la Riva, P.; Meneses, C.F.; Olascoaga, J. Bilateral Retrobulbar Optic Neuropathy Associated With Golimumab. Clin. Neuropharmacol. 2017, 40, 149–151. [Google Scholar] [CrossRef]
  125. Nakao, M.; Asano, Y.; Nakamura, K.; Shida, R.; Takahashi, T.; Yoshizaki, A.; Mitsui, A.; Shibata, S.; Araki, M.; Watanabe, R.; et al. The development of chronic inflammatory demyelinating polyneuropathy during adalimumab treatment in a patient with psoriasis vulgaris. Eur. J. Dermatol. 2016, 26, 404–405. [Google Scholar] [CrossRef]
  126. Naruse, H.; Nagashima, Y.; Maekawa, R.; Etoh, T.; Hida, A.; Shimizu, J.; Kaida, K.; Shiio, Y. Successful treatment of infliximab-associated immune-mediated sensory polyradiculopathy with intravenous immunoglobulin. J. Clin. Neurosci. 2013, 20, 1618–1619. [Google Scholar] [CrossRef] [PubMed]
  127. Birnbaum, J.; Bingham, C.O. Non-length-dependent and length-dependent small-fiber neuropathies associated with tumor necrosis factor (TNF)-inhibitor therapy in patients with rheumatoid arthritis: Expanding the spectrum of neurological disease associated with TNF-inhibitors. Semin. Arthritis Rheum. 2014, 43, 638–647. [Google Scholar] [CrossRef] [PubMed]
  128. Rowan, C.R.; Tubridy, N.; Cullen, G. Multifocal Motor Neuropathy Associated with Infliximab. J. Crohn’s Colitis 2015, 9, 1174–1175. [Google Scholar] [CrossRef] [PubMed]
  129. Genç, H.M.; Kutlubay, B.; Sürmeli, R.; Sözen, H.G.; Sözeri, B. Chronic inflammatory demyelinating neuropathy after etanercept therapy in the course of juvenile idiopathic arthritis. Turk. J. Pediatr. 2023, 65, 165–169. [Google Scholar] [CrossRef]
  130. Wong, S.L.; Rajabally, Y.A. Steroid-induced inflammatory neuropathy in a patient on tumor necrosis factor-α antagonist therapy. J. Clin. Neuromuscul. Dis. 2010, 12, 88–90. [Google Scholar] [CrossRef]
  131. Yao, A.; Chan, H.; Macdonell, R.A.L.; Shuey, N.; Khong, J.J. Bilateral facial nerve palsies secondary to chronic inflammatory demyelinating polyneuropathy following adalimumab treatment. Clin. Neurol. Neurosurg. 2018, 164, 64–66. [Google Scholar] [CrossRef]
  132. Lv, Z.; Zhang, X.; Wu, L. Infliximab-induced seizures in a patient with Crohn’s disease: A case report. BMC Gastroenterol. 2021, 21, 193. [Google Scholar] [CrossRef]
  133. Brigo, F.; Bongiovanni, L.G.; Cerini, R.; Manganotti, P.; Storti, M.; Fiaschi, A. Infliximab-related seizures: A first case study. Epileptic Disord. 2011, 13, 214–217. [Google Scholar] [CrossRef]
  134. Soubrier, M.; Haïk, S.; Hauw, J.-J.; Corvol, J.C.; Lyon-Caen, O.; Dougados, M. Creutzfeldt-Jakob disease in a patient treated by etanercept for rheumatoid arthritis (RA): Just a coincidence? Jt. Bone Spine 2010, 77, 174–175. [Google Scholar] [CrossRef]
  135. de Oliveira, K.F.; Martins, G.A.; Galvão, K.R.F.; Kurizky, P.S. Lewis-Sumner syndrome associated with infliximab therapy for psoriasis. An. Bras. Dermatol. 2017, 92, 156–157. [Google Scholar] [CrossRef]
  136. Cirillo, G.; Todisco, V.; Tedeschi, G. Lewis–Sumner syndrome associated with infliximab therapy in ulcerative colitis. Neurol. Sci. 2016, 37, 1005–1008. [Google Scholar] [CrossRef] [PubMed]
  137. Nancey, S.; Bouhour, F.; Boschetti, G.; Magnier, C.; Gonneau, P.-M.; Souquet, J.-C.; Kaiserlian, D.; Flourie, B. Lewis and Sumner syndrome following infliximab treatment in Crohn’s disease: A report of 2 cases. Inflamm. Bowel Dis. 2010, 16, 1450–1453. [Google Scholar] [CrossRef] [PubMed]
  138. Kurmann, P.T.; Van Linthoudt, D.; So, A.K.-L. Miller–Fisher syndrome in a patient with rheumatoid arthritis treated with adalimumab. Clin. Rheumatol. 2009, 28, 93–94. [Google Scholar] [CrossRef]
  139. Ratnarajan, G.; Thompson, A.; Dodridge, C.; Parry, A.; Elston, J. Novel Variant of Miller Fisher Syndrome Occurring With Tumor Necrosis Factor α Antagonist Therapy. JAMA Neurol. 2015, 72, 1377. [Google Scholar] [CrossRef] [PubMed]
  140. Gaudio, A.; Corrado, A.; Santoro, N.; Maruotti, N.; Cantatore, F.P. Melkersson-Rosenthal Syndrome in a Patient with Psoriatic Arthritis Receiving Etanercept. Int. J. Immunopathol. Pharmacol. 2013, 26, 229–233. [Google Scholar] [CrossRef]
  141. Cançado, G.G.L.; Vilela, E.G. Guillain–Barré syndrome during adalimumab therapy for Crohn’s disease: Coincidence or consequence? Scand. J. Gastroenterol. 2017, 52, 473–476. [Google Scholar] [CrossRef]
  142. de Natividade, N.B.; Felix, P.A.O.; Lerer, C. Guillain-Barré syndrome in a patient on adalimumab for the treatment of psoriasis. An. Bras. Dermatol. 2017, 92, 85–87. [Google Scholar] [CrossRef]
  143. Alvarez-Lario, B.; Prieto-Tejedo, R.; Colazo-Burlato, M.; Macarrón-Vicente, J. Severe Guillain–Barré syndrome in a patient receiving anti-TNF therapy. Consequence or coincidence. A case-based review. Clin. Rheumatol. 2013, 32, 1407–1412. [Google Scholar] [CrossRef]
  144. Patwala, K.; Crump, N.; De Cruz, P. Guillain-Barré syndrome in association with antitumour necrosis factor therapy: A case of mistaken identity. BMJ Case Rep. 2017, 2017, bcr-2017. [Google Scholar] [CrossRef]
  145. Bouchra, A.; Benbouazza, K.; Hajjaj-Hassouni, N. Guillain–Barre in a patient with ankylosing spondylitis secondary to ulcerative colitis on infliximab therapy. Clin. Rheumatol. 2009, 28, 53–55. [Google Scholar] [CrossRef]
  146. Cesarini, M.; Angelucci, E.; Foglietta, T.; Vernia, P. Guillain–Barrè syndrome after treatment with human anti-tumor necrosis factorα (adalimumab) in a Crohn’s disease patient: Case report and literature review. J. Crohn’s Colitis 2011, 5, 619–622. [Google Scholar] [CrossRef] [PubMed]
  147. Lee, J.-H.; Park, J.-S.; Park, D. Successful IVIG treatment without discontinuation of TNF-α blocker in Guillain-Barre syndrome induced by adalimumab in patient with Crohn’s disease. Neurol. Sci. 2018, 39, 595–598. [Google Scholar] [CrossRef] [PubMed]
  148. Psarelis, S.; Hajineocli, A.P.D.; Hadjicosta, E.; Elliott, H.S.A.; Johnson, P. Is secukinumab a safe alternative treatment for ankylosing spondylitis with Guillain Barré syndrome after anti-TNF-α treatment? Case report and literature review. Clin. Rheumatol. 2017, 36, 1197–1199. [Google Scholar] [CrossRef]
  149. Stacy, D.; Johnson, R.; Pendleton, J. Demyelinating Disease Associated with Use of Etanercept in Patients with Seronegative Spondyloarthropathies. J. Rheumatol. 2008, 35, 1469–1471. [Google Scholar]
  150. Morís, G. Inflammatory bowel disease: An increased risk factor for neurologic complications. World J. Gastroenterol. 2014, 20, 1228. [Google Scholar] [CrossRef] [PubMed]
  151. Solomon, A.J.; Spain, R.I.; Kruer, M.C.; Bourdette, D. Inflammatory neurological disease in patients treated with tumor necrosis factor alpha inhibitors. Mult. Scler. J. 2011, 17, 1472–1487. [Google Scholar] [CrossRef] [PubMed]
  152. Kunchok, A.; Aksamit, A.J.; Davis, J.M.; Kantarci, O.H.; Keegan, B.M.; Pittock, S.J.; Weinshenker, B.G.; McKeon, A. Association Between Tumor Necrosis Factor Inhibitor Exposure and Inflammatory Central Nervous System Events. JAMA Neurol. 2020, 77, 937. [Google Scholar] [CrossRef]
  153. Garg, N.; Woltjer, R.; Hamilton, B.; Neuwelt, E.A.; Rosenbaum, J.T. A 67-Year-Old Woman Receiving Tumor Necrosis Factor α Inhibitor Therapy Presenting With Neurologic Dysfunction. Arthritis Care Res. 2013, 65, 658–665. [Google Scholar] [CrossRef]
  154. van der Bijl, A.E.; Emmer, B.J.; Breedveld, F.C.; Middelkoop, H.A.M.; Jurgens, C.K.; van Buchem, M.A.; Huizinga, T.W.J.; van der Grond, J. Advanced magnetic resonance imaging of the brain in patients treated with TNF-alpha blocking agents. Clin. Exp. Rheumatol. 2007, 25, 301–304. [Google Scholar]
  155. Baumer, F.M.; Ouahed, J.; Verhave, M.; Rivkin, M.J. Fatal Central Nervous System Disease Following First Infliximab Infusion in a Child With Inflammatory Bowel Disease. Pediatr. Neurol. 2016, 57, 91–94. [Google Scholar] [CrossRef]
  156. Bando, Y. Mechanism of demyelination and remyelination in multiple sclerosis. Clin. Exp. Neuroimmunol. 2020, 11, 14–21. [Google Scholar] [CrossRef]
  157. Theodoridou, A.; Settas, L. Demyelination in rheumatic diseases. Postgrad. Med. J. 2008, 84, 127–132. [Google Scholar] [CrossRef] [PubMed]
  158. Atzeni, F.; Talotta, R.; Masala, I.F.; Gerardi, M.C.; Casale, R.; Sarzi-Puttini, P. Central nervous system involvement in rheumatoid arthritis patients and the potential implications of using biological agents. Best Pract. Res. Clin. Rheumatol. 2018, 32, 500–510. [Google Scholar] [CrossRef] [PubMed]
  159. Atmaca, M.M.; Altiokka Uzun, G.; Shugaiv, E.; Kurtuncu, M.; Eraksoy, M. Association of Demyelinating and Inflammatory Bowel Diseases: A Case Series and Overview of the Literature. Noro Psikiyatr. Ars. 2015, 52, 315–318. [Google Scholar] [CrossRef]
  160. Nowowiejska, J.; Baran, A.; Flisiak, I. Psoriasis and neurodegenerative diseases—A review. Front. Mol. Neurosci. 2022, 15, 917751. [Google Scholar] [CrossRef]
  161. Sofat, N.; Malik, O.; Higgens, C.S. Neurological involvement in patients with rheumatic disease. QJM Int. J. Med. 2006, 99, 69–79. [Google Scholar] [CrossRef]
  162. Khedr, E.M.; Rashad, S.M.; Hamed, S.A.; El-Zharaa, F.; Abdalla, A.K.H. Neurological complications of ankylosing spondylitis: Neurophysiological assessment. Rheumatol. Int. 2009, 29, 1031–1040. [Google Scholar] [CrossRef]
  163. Jang, D.; Lee, A.-H.; Shin, H.-Y.; Song, H.-R.; Park, J.-H.; Kang, T.-B.; Lee, S.-R.; Yang, S.-H. The Role of Tumor Necrosis Factor Alpha (TNF-α) in Autoimmune Disease and Current TNF-α Inhibitors in Therapeutics. Int. J. Mol. Sci. 2021, 22, 2719. [Google Scholar] [CrossRef]
  164. Zahid, M.; Busmail, A.; Penumetcha, S.S.; Ahluwalia, S.; Irfan, R.; Khan, S.A.; Rohit Reddy, S.; Vasquez Lopez, M.E.; Mohammed, L. Tumor Necrosis Factor Alpha Blockade and Multiple Sclerosis: Exploring New Avenues. Cureus 2021, 13, e18847. [Google Scholar] [CrossRef]
  165. Ortí-Casañ, N.; Boerema, A.S.; Köpke, K.; Ebskamp, A.; Keijser, J.; Zhang, Y.; Chen, T.; Dolga, A.M.; Broersen, K.; Fischer, R.; et al. The TNFR1 antagonist Atrosimab reduces neuronal loss, glial activation and memory deficits in an acute mouse model of neurodegeneration. Sci. Rep. 2023, 13, 10622. [Google Scholar] [CrossRef]
  166. Fischer, R.; Kontermann, R.E.; Pfizenmaier, K. Selective Targeting of TNF Receptors as a Novel Therapeutic Approach. Front. Cell Dev. Biol. 2020, 8, 401. [Google Scholar] [CrossRef] [PubMed]
  167. Kaltsonoudis, E.; Voulgari, P.V.; Konitsiotis, S.; Drosos, A.A. Demyelination and other neurological adverse events after anti-TNF therapy. Autoimmun. Rev. 2014, 13, 54–58. [Google Scholar] [CrossRef] [PubMed]
  168. Ye, C.; Geng, Z.; Li, S.; Yu, F. Etanercept for Ankylosing Spondylitis With Coexisting Demyelinating Myelitis. Am. J. Ther. 2019, 26, e629–e631. [Google Scholar] [CrossRef]
  169. Cisternas, M.; Gutiérrez, M.; Jacobelli, S. Successful rechallenge with anti–tumor necrosis factor α for psoriatic arthritis after development of demyelinating nervous system disease during initial treatment: Comment on the article by Mohan et al. Arthritis Rheum. 2002, 46, 3107–3108. [Google Scholar] [CrossRef] [PubMed]
Figure 1. TNF-α mechanism of action. For details, see the text above. Created with biorender.com.
Figure 1. TNF-α mechanism of action. For details, see the text above. Created with biorender.com.
Medicina 60 01409 g001
Figure 2. Percentage distribution of TNF-α inhibitors used in various diseases.
Figure 2. Percentage distribution of TNF-α inhibitors used in various diseases.
Medicina 60 01409 g002
Figure 3. Diagnosed conditions after treatment with TNF-α inhibitors: autoimmune encephalitis (purple), CNS demyelination (light purple), inflammatory demyelination (pink), meningitis (yellow–green), MRS (cyan), multifocal motor neuropathy (red), myelopathy (light blue), non-demyelinating CNS inflammation (light green), peripheral demyelination (grey); neurodegenerative diseases (green); seizures (dark red); neurosarcoidosis (yellow); LSS (blue); Henoch–Schönlein purpura (dark yellow). INFL (infliximab), GOL (golimumab), ETA (etanercept), CZP (certolizumab), ADA (adalimumab).
Figure 3. Diagnosed conditions after treatment with TNF-α inhibitors: autoimmune encephalitis (purple), CNS demyelination (light purple), inflammatory demyelination (pink), meningitis (yellow–green), MRS (cyan), multifocal motor neuropathy (red), myelopathy (light blue), non-demyelinating CNS inflammation (light green), peripheral demyelination (grey); neurodegenerative diseases (green); seizures (dark red); neurosarcoidosis (yellow); LSS (blue); Henoch–Schönlein purpura (dark yellow). INFL (infliximab), GOL (golimumab), ETA (etanercept), CZP (certolizumab), ADA (adalimumab).
Medicina 60 01409 g003
Figure 4. Primary disease outcome after treatment with TNF-α inhibitor: mild improvement (purple), no improvement (light purple), not reported (pink), partial resolution (yellow–green), significant improvement (cyan), significant resolution (gray), worsening neurological symptoms (green). INFL (infliximab), GOL (golimumab), ETA (etanercept), CZP (certolizumab), ADA (adalimumab).
Figure 4. Primary disease outcome after treatment with TNF-α inhibitor: mild improvement (purple), no improvement (light purple), not reported (pink), partial resolution (yellow–green), significant improvement (cyan), significant resolution (gray), worsening neurological symptoms (green). INFL (infliximab), GOL (golimumab), ETA (etanercept), CZP (certolizumab), ADA (adalimumab).
Medicina 60 01409 g004
Figure 5. MS pathology. T cells, macrophages, and antigen-presenting cells (APCs) enter the central nervous system (CNS) and secrete cytokines that damage the oligodendrocytes, cells that produce myelin in the CNS. B cells can produce myelin-specific antibodies which will lead to complement cascade activation and membrane–attack complex formation that further damages the oligodendrocytes. A new hypothesis suggests that anti-myelin oligodendrocyte glycoprotein (anti-MOG) autoantibody will induce stress in the oligodendrocyte endoplasmic reticulum (ER). This event will induce morphological abnormalities in oligodendrocytes and demyelination in the broad sense. Moreover, oligodendrocytes secrete Kallikrein 6 (KLK6) and matrix metalloprotease (MMP)-9, which will damage the blood–brain barrier (BBB) and exacerbate the pathology of multiple sclerosis [156]. Created with biorender.com.
Figure 5. MS pathology. T cells, macrophages, and antigen-presenting cells (APCs) enter the central nervous system (CNS) and secrete cytokines that damage the oligodendrocytes, cells that produce myelin in the CNS. B cells can produce myelin-specific antibodies which will lead to complement cascade activation and membrane–attack complex formation that further damages the oligodendrocytes. A new hypothesis suggests that anti-myelin oligodendrocyte glycoprotein (anti-MOG) autoantibody will induce stress in the oligodendrocyte endoplasmic reticulum (ER). This event will induce morphological abnormalities in oligodendrocytes and demyelination in the broad sense. Moreover, oligodendrocytes secrete Kallikrein 6 (KLK6) and matrix metalloprotease (MMP)-9, which will damage the blood–brain barrier (BBB) and exacerbate the pathology of multiple sclerosis [156]. Created with biorender.com.
Medicina 60 01409 g005
Figure 6. The role of TNF-α in PSO, RA, PsA, and uveitis. In RA, macrophage and Th1 cells secrete TNF-α, activating the synovial fibroblasts, which leads to excessive production of cathepsins and matrix metalloproteinases (MMPs). This overproduction will result in cartilage and bone destruction and consequently joint erosion. In PSO, the stressed keratinocytes secrete TNF-α, which activates dendritic cells (DCs), inducing the activation of Th1 cells and Th17 cells. The activated T cells secrete TNF-α and IL-17, which cause epidermal changes and keratinocyte hyperproliferation. Activation of DCs by TNF-α secreted by macrophages induces migration of T cells and inflammation responsible for uveitis. In PsA, macrophages secrete TNF-α, which activates in parallel the synovial fibroblasts and DCs. DC activation will further activate Th17, which will produce keratinocyte hyperproliferation and epidermal changes and activate osteoclasts. Created with biorender.com.
Figure 6. The role of TNF-α in PSO, RA, PsA, and uveitis. In RA, macrophage and Th1 cells secrete TNF-α, activating the synovial fibroblasts, which leads to excessive production of cathepsins and matrix metalloproteinases (MMPs). This overproduction will result in cartilage and bone destruction and consequently joint erosion. In PSO, the stressed keratinocytes secrete TNF-α, which activates dendritic cells (DCs), inducing the activation of Th1 cells and Th17 cells. The activated T cells secrete TNF-α and IL-17, which cause epidermal changes and keratinocyte hyperproliferation. Activation of DCs by TNF-α secreted by macrophages induces migration of T cells and inflammation responsible for uveitis. In PsA, macrophages secrete TNF-α, which activates in parallel the synovial fibroblasts and DCs. DC activation will further activate Th17, which will produce keratinocyte hyperproliferation and epidermal changes and activate osteoclasts. Created with biorender.com.
Medicina 60 01409 g006
Table 2. Details on the neurologic events after treatment with TNF-α inhibitors.
Table 2. Details on the neurologic events after treatment with TNF-α inhibitors.
Primary DiseaseTNF-α InhibitorAge/SexFamily History of Neurological DiseasesNeurological Symptoms/Diagnosed ConditionsTherapeutic MeasuresOutcomeRef.
1.RAINFLNot reported/MNot reportedALSDiscontinuation
Riluzole
Progressive ALS[59]
2.ASINFL55/MNot reportedALSDiscontinuation
Riluzole, alphatocopherol
Progressive ALS; death by respiratory insufficiency[60]
3.RAINFL65/FNot reportedALSDiscontinuation
Riluzole not tolerated
Progressive ALS; death by respiratory insufficiency[61]
4.CDADA53/FNot reportedDemyelinating lesions/autoimmune encephalitisDiscontinuation
Intravenous steroids, immunoglobulin
Resolution[62]
5.ASADA30/FNot reportedDemyelinating lesions/autoimmune encephalitis (Bickerstaff
brainstem encephalitis)
Discontinuation
Methylprednisolone, intravenous gamma globulin
(IVIg), rituximab
Partial resolution[63]
6.CDADA20/FNot reportedPeripheral neuropathy/Henoch–Schönlein purpura with neurological involvementDiscontinuation
ADA replaced with ustekinumab/methylprednisolone
Resolution[64]
7.CDINFL64/FNeurofibromatosisAcute neuropathy, encephalopathyDiscontinuation
Polyvalent immunoglobulin
Partial resolution[65]
8.CDINFL27/FNot reportedNon-demyelinating inflammatory CNS events/anti-NMDAR
encephalitis
INFL replaced with ustekinumab
Rituximab
Resolution[66]
9.RAINFL72/MNot reportedDemyelinating progressive multifocal leukoencephalopathyDiscontinuationPersistent neurologic and cognitive deficits[67]
10.CDINFL45/FNot reportedAcute disseminated encephalomyelitisDiscontinuation
Methylprednisolone
Resolution[68]
11.RAADA66/FNot reportedDemyelinating progressive multifocal leukoencephalopathyDiscontinuationWorsening neurological symptoms[67]
12.RAADA66/MNot reportedDemyelinating leukoencephalopathyDiscontinuation
Methylprednisolone
Meloxicam
Partial resolution[69]
13.ASGOL45/FNot reportedDemyelinating posterior reversible encephalopathy syndromeDiscontinuation, anti-convulsant therapy (levetiracetam)Resolution[70]
14.RAETA74/FNot reportedDemyelinating progressive multifocal leukoencephalopathyDiscontinuation
Supportive therapy
Slight improvement[71]
15.RAETA47/FNot reportedLeptomeningitisDiscontinuation
Methylprednisolone, azathioprine, levetiracetam, valproic acid.
Significant resolution[72]
16.RAADA77/MNot reportedHypertrophic pachymeningitisDiscontinuation, PrednisoneResolution[73]
17.JIAADA18/FNot reportedAseptic meningitisDiscontinuation
Methylprednisolone
Resolution[74]
18.RAADA52/FNot reportedAcute
myelopathy/paraplegia
Discontinuation
Methylprednisolone (oral/i.v)
Partial resolution[75]
19.CDADA52/FChronic myelitisPartial resolution
20.RAADA42/MChronic myelitisNo response
21.RA
Lupus
CZP54/FChronic myelitisPartial resolution
22.Blau syndromeETA13/MNot reportedTransverse myelitisDiscontinuation
Methylprednisolone, prednisone
Resolution[76]
23.Behcet’s diseaseGOL34/MNot reportedDemyelinating lesionsDiscontinuation
Steroid therapy
Resolution[77]
24.Behcet’s diseaseGOL51/FNot reportedMultifocal myelitisDiscontinuation
Steroid therapy
Resolution[78]
25.CDINFL40/MNot reportedExtensive longitudinal myelopathyDiscontinuation
Methylprednisolone, followed by oral prednisolone
Slight improvement[79]
26.ASADA
ETA
50/MNot reportedRapidly progressive dementiaDiscontinuationResolution[80]
27.RAADA40/FNoneCNS demyelinationDiscontinuation
Methylprednisolone
Resolution[81]
28.PSOADA36/FNoneCNS demyelinationDiscontinuationResolution[82]
29.PSOADA37/FNoneCNS demyelinationDiscontinuation
Intravenous immunoglobulin, oral corticosteroids
Resolution[83]
30.RAADA39/FNot reportedChronic demyelinating neuropathyDiscontinuationResolution[84]
31.CDADA34/FNoneInflammatory demyelinationDiscontinuation
Steroid therapy
Slight improvement[85]
32.HSADA29/MNot reportedDemyelinating radiologically isolated syndromeDiscontinuationResolution[86]
33.JIAADA14/MNot reportedCNS active inflammatory process (no demyelination lesions)Discontinuation
High-dose corticosteroids
Partial resolution[87]
34.PSOADA51/FNoneMSDiscontinuation
high-dose corticosteroids
Partial resolution[88]
35.ASETA34/MNoneCNS demyelinationDiscontinuationSlight improvement[89]
36.ASETA26/FNot reportedMOGADDiscontinuation
Methylprednisolone
Significant improvement[90]
37.ASETA44/FNot reportedTumefactive CNS inflammatory demyelinationDiscontinuation
High-dose steroids
Significant improvement[91]
38.ASETA36/MMS (brother)Acute transverse myelitisDiscontinuation
Methylprednisolone
Resolution[92]
39.Juvenile RAETA21/FNoneMS
Optic neuritis
Discontinuation
Methylprednisolone
Partial resolution[93]
40.PSOETA55/MNoneMSDiscontinuation
Methylprednisolone
No improvement[94]
41.ASADA44/MNoneDemyelinating brain lesionsDiscontinuationNot reported
42.RAETA26/FNot reportedMSNot reportedNot reported[95]
43.ASGOL41/MNoneFulminant CNS demyelinationDiscontinuation
Steroid therapy
Plasmapheresis
Resolution[96]
44.PSOINFL47/FMS (sister)MSDiscontinuation
Steroid therapy
Partial resolution[97]
45.PSOINFL27/FNoneCNS demyelinationDiscontinuation
Steroid therapy
Rituximab
Partial resolution[98]
46.CDINFL46/MNoneInflammatory demyelinating polyneuropachronicthDiscontinuation
Intravenous human immunoglobulin
Resolution[99]
47.CD
AS
INFL18/FNot reportedCNS demyelinationDiscontinuationNo improvement[100]
48.AS
Uveitis
INFL38/MNot reportedDemyelinating brain lesionsDiscontinuationResolution[101]
49.RAINFL56/MNot reportedCNS demyelinationDiscontinuation
High-dose dexamethasone
Resolution[102]
50.66/FNot reportedDiscontinuation
Oral prednisone
Significant improvement
51.CDINFL19/FNoneCNS demyelinationDiscontinuationResolution[103]
52.Uveitis
JIA
ADA21/FNot reportedMSDiscontinuation
Cladribine
Resolution[38]
53.PSOADA56/FNot reportedMS
aseptic meningitis
DiscontinuationResolution[104]
54.RAADA68/FNoneMSDiscontinuationResolution[105]
55.Autoimmune UveitisADA23/MMS (family)MSDiscontinuation
Methylprednisolone, oral prednisolone
Resolution[39]
56.ASADA36/MNoneMSDiscontinuation
Methylprednisolone, oral prednisolone
Resolution[106]
57.CDINFL47/FNoneMSDiscontinuationResolution[107]
58.UCINFL35/FMS (family)MSDiscontinuationNot reported[108]
59.ASINFL48/MNot reportedMSDiscontinuationResolution[109]
60.PSOADA41/MCNS demyelinationDiscontinuationResolution
61.PSOETA38/FMSDiscontinuation
Interferon beta-1a
Not reported
62.RAADA64/FMSDiscontinuationNot reported
63.ASINFL34/FClinical isolated syndromeDiscontinuationResolution
64.PSOINFL57/MPeripheral demyelinating
neuropathy
DiscontinuationResolution
65.PSOETA48/MNot reportedMSDiscontinuationResolution[110]
66.PSOINFL62/FNot reportedOptic neuritisDiscontinuation
Methylprednisolone
No improvement[111]
67.PSOINFL56/FNot reportedCNS demyelinationDiscontinuation
Methylprednisolone
Significant improvement[112]
68.PSOCZP44/MNot reportedCranial nerve III demyelinationDiscontinuation
methylprednisolone
Resolution[113]
69.RAETA64/FNot reportedMyelitis
optic neuritis
Discontinuation
Methylprednisolone
Resolution[114]
70.CDADA27/FNot reportedInternuclear ophthalmoplegiaDiscontinuation
Steroid therapy
Resolution[115]
71.RAINFL53/FNot reportedOptic neuritisDiscontinuation
Steroid therapy
Resolution[116]
72.RAETA40/FNot reportedNeurosarcoidosisDiscontinuation
Methylprednisolone, oral prednisolone
Partial resolution[117]
73.RA
Uveitis
ETA33/FNot reportedNeurosarcoidosisETA replaced with INFL
Steroid therapy
Significant improvement[118]
74.RAINFL50/FNonePolineuropathyDiscontinuation
Intravenous gammaglobulin
Slight improvement[119]
75.85/FMild sensory lossDiscontinuation
Steroid therapy
Partial resolution
76.68/FNot reportedDiscontinuationSlight improvement
77.PSOADA65/FNot reportedAcute bilateral symmetric phrenic neuropathyDiscontinuation
Oxygen
Resolution[120]
78.RAETA45/FNot reportedChronic inflammatory demyelinating polyneuropathyDiscontinuation
Intravenous gammaglobulin
Partial resolution[121]
79.INFL49/MNot reported
80.PSOADA53/FGBSChronic inflammatory demyelinating polyradiculoneuropathyDiscontinuation
Intravenous immunoglobulin
Resolution[122]
81.RAADA52/MIdiopathic Parkinson’s diseaseDADS
neuropathy
DiscontinuationResolution[123]
82.ASGOL48/MALS (family)Bilateral retrobulbar optic neuropathyDiscontinuation
Methylprednisolone, oral prednisolone
No improvement[124]
83.PSOADA42/MNot reportedChronic inflammatory demyelinating polyneuropathyDiscontinuation
intravenous immunoglobulin
Resolution[125]
84.PSOINFL64/MNot reportedSensory polyradiculopathyDiscontinuation
Intravenous immunoglobulin
Resolution[126]
85.RAINFL32/FNot reportedSmall-fiber neuropathiesDiscontinuationPartial resolution[127]
86.73/FNot reportedDiscontinuation
Symptomatic treatment
Partial resolution
87.55/FNot reportedDiscontinuationSignificant improvement
88.ASINFL54/MNot reportedMMNCBDiscontinuationSignificant improvement[128]
89.UCINFL30/MNoneMultifocal motor neuropathyDiscontinuation
Intravenous immunoglobulin
Resolution[128]
90.JIAETA12/FNot reportedChronic inflammatory demyelinating neuropathyDiscontinuation
Intravenous immunoglobulins, steroids, plasma exchange
Limited response[129]
91.RAADA44/FNot reportedInflammatory demyelinating neuropathyDiscontinuation
Intravenous immunoglobulin
Partial resolution[130]
92.CDADA37/MNot reportedChronic inflammatory demyelinating neuropathyDiscontinuation
Intravenous immunoglobulins, steroids
Significant improvement[131]
93.CDINFL60/FNoneSeizuresDiscontinuationResolution[132]
94.CDINFL74/MNot reportedSeizuresDiscontinuationResolution[133]
95.RAETA74/FNot reportedCJDDiscontinuationResolution[134]
96.PSOINFL40/MNot reportedLSSDiscontinuation
Intravenous immunoglobulin
Partial resolution[135]
97.UCINFL44/MNoneLSSDiscontinuation
Intravenous immunoglobulin
Partial resolution[136]
98.CDINFL24/FNot reportedLSSDiscontinuation
Intravenous immunoglobulin
Resolution[137]
99.35/FNot reported
100.RAINFL77/FNot reportedMFSDiscontinuation
High-dose steroids, gabapentine
Slow improvement[138]
101.UCINFL43/FNot reportedMFSDiscontinuationResolution[139]
102.PSOETA42/FNot reportedMFSDiscontinuationSlow improvement[140]
103.CDADA64/MNot reportedGBSDiscontinuationResolution[141]
104.PSOADA45/MNot reportedGBSDiscontinuationPartial resolution[142]
105.RAADA50/FNot reportedGBSDiscontinuation
Prednisone, gabapentin
Partial resolution[143]
106.CDADA37/MNot reportedGBSDiscontinuation
Intravenous immunoglobulin
Partial resolution[144]
107.UC
AS
INFL47/FNot reportedGBSDiscontinuation
intravenous immunoglobulin
Resolution[145]
108.CDADA71/MNot reportedGBSDiscontinuation
Intravenous immunoglobulin, Methylprednisolone, plasmapheresis
Partial resolution[146]
109.CDADA33/FNot reportedGBSContinuation of ADA treatment
Intravenous immunoglobulin
Significant improvement[147]
110.ASETA50/MNot reportedGBSDiscontinuation
Intravenous immunoglobulin, plasmapheresis
Resolution[148]
111.PSOETA53/MNoneMSDiscontinuation
Interferonβ
Baclofen
Significant improvement[149]
112.PSO42/MNoneMSDiscontinuationSignificant improvement
113.AS51/FNot reported DiscontinuationMild improvement
INFL = infliximab, GOL = golimumab, ETA = etanercept, CZP = certolizumab, ADA = adalimumab, RA = rheumatoid arthritis, JIA = juvenile idiopathic arthritis, HS = hidradenitis suppurativa, AS = ankylosing spondylitis, CD = Crohn’s disease, UC = ulcerative colitis; MS = multiple sclerosis, GBS = Guillain–Barré syndrome, MFS = Miller–Fisher syndrome, LSS = lumbar spinal stenosis, CJD = Creutzfeldt–Jakob disease, MMNCB = multifocal motor neuropathy with conduction blocks, DADS = distal acquired demyelinating symmetric neuropathy, MOGAD = myelin oligodendrocyte glycoprotein antibody disease, ALS = amyotrophic lateral sclerosis. Discontinuation = discontinuation of the TNF-α inhibitor therapy.
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

Gogulescu, A.; Blidisel, A.; Soica, C.; Mioc, A.; Voicu, A.; Jojic, A.; Voicu, M.; Banciu, C. Neurological Side Effects of TNF-α Inhibitors Revisited: A Review of Case Reports. Medicina 2024, 60, 1409. https://doi.org/10.3390/medicina60091409

AMA Style

Gogulescu A, Blidisel A, Soica C, Mioc A, Voicu A, Jojic A, Voicu M, Banciu C. Neurological Side Effects of TNF-α Inhibitors Revisited: A Review of Case Reports. Medicina. 2024; 60(9):1409. https://doi.org/10.3390/medicina60091409

Chicago/Turabian Style

Gogulescu, Armand, Alexandru Blidisel, Codruta Soica, Alexandra Mioc, Adrian Voicu, Alina Jojic, Mirela Voicu, and Christian Banciu. 2024. "Neurological Side Effects of TNF-α Inhibitors Revisited: A Review of Case Reports" Medicina 60, no. 9: 1409. https://doi.org/10.3390/medicina60091409

Article Metrics

Back to TopTop