Abstract
The past decades have yielded major therapeutic advances in many autoimmune conditions – such as multiple sclerosis (MS) – and thus ushered in a new era of more targeted and increasingly potent immunotherapies. Yet this growing arsenal of therapeutic immune interventions has also rendered therapy much more challenging for the attending physician, especially when treating patients with more than one autoimmune condition. Importantly, some therapeutic strategies are either approved for several autoimmune disorders or may be repurposed for other conditions, therefore opening new curative possibilities in related fields. In this article, we especially focus on frequent and therapeutically relevant concomitant autoimmune conditions faced by neurologists when treating patients with MS, namely psoriasis, rheumatoid arthritis and inflammatory bowel diseases. We provide an overview of the available disease-modifying therapies, highlight possible contraindications, show pathophysiological overlaps and finally present which therapeutics can be utilized as a combinatory treatment, in order to ‘kill two birds with one stone’.
Keywords: immunotherapy, inflammatory bowel disease, multiple sclerosis, psoriasis, rheumatoid arthritis
Introduction
In the past decades, treatment of autoimmune conditions such as multiple sclerosis (MS) has experienced significant changes, resulting from major advances in disease-modifying therapies (DMTs). However, considering the growing armamentarium of immunomodulatory drugs, selecting the right therapeutic agent for an individual patient has become more challenging. Genome-wide association studies have identified hundreds of shared susceptibility loci for autoimmune diseases,1 and it is not uncommon for patients to have more than one autoimmune disorder. While some of these patients may present therapeutic challenges, it also creates the opportunity to utilize elegantly specific immunosuppressants targeting two autoimmune conditions simultaneously and consequently ‘kill two birds with one stone’. MS is associated with several other auto-inflammatory conditions, which share not only certain susceptibility genes, but also general pathophysiological principles. For example, psoriasis (PsO) – like MS – is a largely T-cell driven disease with the interleukin (IL)23/T helper (Th)17-axis being critically involved in the pathology of both conditions.2,3 These immunological interfaces can be used to repurpose DMTs that were initially developed for other inflammatory disorders and tailor crossover therapies for the individual patient, as shown for the anti-IL17 antibody secukinumab4 or sphingosine-1-phosphate receptor (S1PR) modulators such as ozanimod.5 In contrast, other DMTs may exacerbate pre-existing autoimmune diseases or even trigger their onset. For instance, tumor necrosis factor alpha (TNFα) blockers are known potentially to induce or worsen demyelinating conditions.6–8 Thus, it is important to shed a light on the ever-growing jungle of immunosuppressants and the possibilities to repurpose these substances for the treatment of other inflammatory comorbidities, such as MS.
Of note, systemic cortisone is the common therapeutic denominator for almost all inflammatory conditions, yet due to its long-term unwanted side effects, cortisone is generally used to induce a state of remission quickly and not as a continuously administered DMT. In fact, many DMTs are used to taper cortisone intake to low levels. Thus, we excluded cortisone from this review and instead exclusively focused on long-term DMTs. Furthermore, we specifically focus on the most common concomitant autoimmune diseases that provide specific challenges for clinicians treating patients with MS, namely PsO, rheumatoid arthritis (RA), and inflammatory bowel diseases [IBDs: ulcerative colitis (UC) and Crohn’s disease (CD)]. Thus, the aim of this review is to offer an overview of the available therapeutic tools across those diseases and to propose pragmatic treatment approaches for the selection of potential disease-overlapping substances, when facing patients with MS and coexisting autoimmune disorders.
Common immunosuppressants
Azathioprine (AZA)
Azathioprine (AZA) and its active metabolite 6-mercaptopurine (6-MP) are antimetabolites, which block the de novo synthesis of nucleobases and thus affect rapidly dividing lymphocytes. AZA is approved by the European Medicines Agency (EMA) and the US Food and Drug Administration (FDA) for many inflammatory conditions, including RA9 (Table 1). In IBD, AZA can be utilized in combination with other therapeutics (like TNFα blockers) to induce and maintain remission; however, its effect commences with a delay of several months. While AZA is no longer routinely used in MS (despite being approved in some countries, including Germany), it is an established therapeutic option in other demyelinating conditions, such as low-active neuromyelitis optica spectrum disorder (NMOSD).10,11 In clinical studies, AZA has proved non-inferiority12 or even superiority13 to interferon-beta (IFNβ), although the investigated cohorts were relatively small and the number of adverse events (e.g. nausea, vomiting, and lymphopenia) was generally higher, while AZA additionally increases the long-term risk of malignancies.14 Thus, despite its very limited use in MS now, it is a reasonable option in patients with, among other things, comorbid IBD.
Table 1.
FDA approval status of presented disease-modifying therapies in regards to specific autoimmune disorders.
| Autoimmune disorders | |||||||||||||||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| MS | IBD | PsO | RA | SpA | SLE | PSS | SS | GCA | TA | GPA | PAN | EGPA | MPA | BD | SARC | ||
| Disease-modifying therapies | |||||||||||||||||
| CORT | + | + | + | + | + | + | + | + | + | + | + | + | + | + | + | + | |
| MTX | (+) | (+) | + | + | + | (+) | (+) | (+) | (+) | (+) | (+) | (+) | (+) | (+) | (+) | (+) | |
| AZA | (+) | (+) | (+) | + | (+) | (+) | (+) | (+) | (+) | − | (+) | − | (+) | (+) | (+) | (+) | |
| MMF | − | (+) | (+) | (+) | − | (+) | (+) | (+) | − | − | − | − | − | − | (+) | − | |
| 5-ASA | − | + | (+) | + | (+) | (−) | − | − | − | − | − | − | − | − | − | − | |
| HCQ | − | (+) | − | + | (+) | + | (+) | (+) | − | (+) | − | (+) | − | − | − | (+) | |
| CYA/TAC | (+) | (+) | + | + | (+) | (+) | (+) | (+) | − | (+) | (+) | (+) | (+) | (+) | (+) | (+) | |
| IFN | + | − | − | − | − | − | − | − | − | − | − | − | − | − | (+) | − | |
| TER/LEF | + | − | + | + | (+) | − | − | − | − | − | (+) | − | (+) | (+) | − | − | |
| DMF | + | − | (+) | − | − | − | − | − | − | − | − | − | − | − | − | − | |
| S1PR | + | (+) | − | − | − | − | − | − | − | − | − | − | − | − | − | − | |
| TOC | − | − | − | + | − | (+) | (+) | − | + | (+) | (+) | (+) | (+) | (+) | (+) | − | |
| USTE | − | + | + | − | (+) | − | − | − | − | − | − | − | − | − | − | − | |
| SECU | (+) | − | + | − | + | − | − | − | − | − | − | − | − | − | − | − | |
| Anti-TNFα | − | + | + | + | + | − | − | − | − | (+) | (+) | − | (+) | (+) | (+) | (+) | |
| NATA | + | + | − | − | − | − | − | − | − | − | − | − | − | − | − | − | |
| ALEM | + | − | − | (+) | − | − | − | − | − | − | − | − | − | − | − | − | |
| Anti-CD20 | + | + | − | + | + | + | (+) | (+) | − | − | + | + | + | + | − | − | |
| JAK | − | + | + | + | − | − | − | − | − | − | − | − | − | − | − | − | |
| Abatacept | − | − | − | + | − | − | − | − | − | − | − | − | − | − | − | − | |
| Anakinra | − | − | − | + | − | − | − | − | − | − | − | − | − | − | − | − | |
, FDA approved; (+), used off-label; −, not generally used.
5-ASA, 5-aminosalicylic acid; ALEM, alemtuzumab; Anti-CD20, CD20 antibodies; Anti-TNFα, TNFα antibodies; AZA, azathioprine; BD, Behcet’s disease; CORT, cortisone; CYA/TAC, cyclosporine/tacrolimus; DMF, dimethylfumarate; EGPA, eosinophilic granulomatosis with polyangiitis; FDA, US Food and Drug Administration; GCA, giant cell arteritis; GPA, granulomatosis with polyangiitis; HCQ, hydroxchloroquine; IBD, inflammatory bowel disease; IFN, interferon; JAK, Janus kinase inhibitor; MMF, mycophenolate mofetil; MPA, microscopic polyangiitis; MS, multiple sclerosis; MTX, methotrexate; NATA, natalizumab; PAN, polyarteritis nodosa; PsO, psoriasis; PSS, primary systemic sclerosis; RA, rheumatoid arthritis; S1PR, sphingosin-1-phosphate receptor modulator; SARC, sarcoidosis; SEC, secukinumab; SLE, systemic lupus erythematosus; SpA, spondylo arthritis; SS, Sjögren’s syndrome; TA, Takayasu arteritis; TER/LEF, teriflunomide/leflunomide; TOC, tocilizumab; USTE, ustekinumab.
Cyclosporine A and tacrolimus
Cyclosporine A (CYA) and tacrolimus (TAC) are inhibitors of calcineurin, an enzyme involved in the transcription of IL-2 and thus T-cell activation and proliferation. Both substances have a broad operational field, including RA,15 IBD16,17 and PsO15,18 (Table 1). Especially in diseases with cutaneous manifestations, CYA and TAC can also be applied topically. CYA has been shown to reduce the relapse rate in relapsing–remitting multiple sclerosis (RRMS),19,20 and there are also promising case reports and small trials for TAC in MS.21 However, with more potent and targeted DMTs in hand, both substances are not first line treatment options for IBD/RA/PsO patients with comorbid MS.
(Hydroxy-)chloroquine (HCQ)
The anti-malarial agent hydroxychloroquine (HCQ) blocks lysosomal enzymes by raising the intra-lysosomal pH, thus hampering the presentation of antigens to CD4+ T cells via major histocompatibility complex (MHC) II. HCQ is in clinical use for several autoimmune conditions including systemic lupus erythematosus (SLE) and RA (Table 1), yet has proved to be less efficient than other disease-modifying antirheumatic drugs (DMARDs).22 So far, there is only experimental evidence [in experimental autoimmune encephalomyelitis (EAE) and cuprizone models] for the efficacy of HCQ in neuroinflammatory conditions.23–25 However, there are two ongoing clinical trials for the application of HCQ in progressive MS (NCT02913157 and NCT03109288); publication of the final results is still pending. Thus, data are currently too limited to recommend HCQ as a treatment option in patients with MS and coexisting autoimmune disorders.
Methotrexate (MTX)
Methotrexate (MTX) is an antimetabolite and competitive inhibitor of folate-dependent enzymes, such as dihydrofolate reductase (DHFR), thereby blocking purine and pyrimidine base biosynthesis necessary for fast (T-) cell proliferation. MTX is generally used for the treatment of RA, IBD, and PsO,15,18 is well tolerated and is thus in rampant use for many autoimmune conditions including vasculitis and SLE (Table 1). Despite some favorable effects on relapse rates and disease progression in early trials,26,27 more potent and targeted DMTs were developed as first line treatment options in MS. However, MTX may serve as a therapy for MS patients with comorbid RA, IBD or PsO.
Mycophenolate mofetil (MMF)
Mycophenolate mofetil (MMF) inhibits the enzyme inosine monophosphate dehydrogenase, which is crucially involved in de novo biosynthesis of purine nucleobases. MMF is relatively specific to lymphocytes, because these cells lack salvage pathways for purine biosynthesis compensating for this deficiency. MMF has been widely used for immunosuppression after organ transplantation and has proved to be effective in various autoimmune conditions including IBD,28 PsO29 and RA.30 In a retrospective analysis conducted by Michel et al.,31 MMF failed to influence positively MS-related disability progression as assessed using the Expanded Disability Status Scale (EDSS), yet significantly reduced relapse rates in MS patients. This was further corroborated in a more recent study on patients with progressive MS.32 Due to limited data on MS and its lack of approval for use in MS, PsO, RA or IBD, MMF might not be among the first choices for a cross-over therapy.
5-Aminosalicylate (5-ASA)
Sulfasalazine consists of sulfapyridine and 5-aminosalicylic acid (5-ASA), which is either absorbed intact or as sulfapyridine. While 5-ASA affects arachidonic acid metabolism and thereby inflammation in IBD, intact sulfasalazine or sulfapyridine – like MTX – have been shown to inhibit folate-dependent enzymes and both appear to be active in RA.22 Thus, 5-ASA is approved for the treatment of IBD and RA33,34 (Table 1). Regarding MS, sulfasalazine has promoted remyelination and suppressed EAE in several animal models of MS,35,36 yet aggravated EAE or completely failed to show beneficial effects in rat EAE models.37,38 In clinical studies, 5-ASA did not reduce EDSS progression39 and there is also a case report of a patient developing MS under a long treatment with sulfasalazine.40 Thus, sulfasalazine may not be considered a suitable therapeutic option for MS, but oral or topical sulfasalazine may serve as an add-on DMT for RA and IBD.
Monoclonal antibodies
Alemtuzumab
Alemtuzumab is a humanized monoclonal antibody targeting and thus depleting CD52-expressing B and T cells. Because of its safety profile, the use of alemtuzumab is reserved for highly active RRMS patients who have not sufficiently responded to other therapies.41 Initially, the efficacy of alemtuzumab was assessed for the treatment of therapy-refractory RA.42 However, because of the induction of profound lymphopenia with a concomitant slow immune reconstitution, the trials were eventually cut short.43 So far, there is only one case report showing a beneficial effect of alemtuzumab in a patient with both chronic plaque PsO and MS;44 there is only limited experimental evidence for the benefit of an anti-CD52 therapy in IBD.45 Thus, a certain subgroup of patients with highly active RRMS plus PsO/RA may profit from a treatment with alemtuzumab.
Anti-CD20 antibodies (rituximab, ocrelizumab, ofatumumab)
Anti-CD20 antibodies target and deplete CD20-expressing B cells. Rituximab has been in clinical use for moderate to severe RA since 2006, but has also been utilized for other autoimmune conditions, including demyelinating disorders and certain types of leukaemia.46,47 There is increasing evidence that T and B cells both play equally important roles in the pathogenesis of MS.48 Moreover, there are several studies and case reports showing the efficacy of rituximab in MS. However, the antibody has not been formally approved for the treatment of RRMS and is thus used off-label to a varying degree with considerable variations across different countries.49,50 Recently, the humanized anti-CD20 antibody ocrelizumab has been shown to reduce disease activity of RRMS and primary progressive multiple sclerosis (PPMS) patients and is the first approved substance for the treatment of PPMS.51,52 Ocrelizumab has also been tested in RA patients, showing a promising safety profile and therapeutic efficacy.53 In IBD and PsO, the reports for CD20 antibodies have not been as promising: an Icelandic study recently reported a six-fold increased risk of developing an IBD in patients treated with rituximab,54 and there are several case reports of patients who developed colitis or PsO during an anti-CD20 treatment with rituximab or ocrelizumab.55–58 Immunity critically relies on the homeostasis of pro and anti-inflammatory stimuli and CD20+ cells have an important regulatory and protective role in the gastrointestinal system59,60 as well as the skin;61 thus B cell depletion may trigger an abnormal B cell-mediated T cell regulation and, as a consequence, PsO or IBD. Thus, CD20 antibodies may be the ideal therapeutic for patients with highly active MS and RA, yet should be carefully considered in MS plus IBD/PsO patients.
Natalizumab
Natalizumab is a humanized IgG4 monoclonal antibody that specifically targets the α4 integrin and thus hampers the invasion of (autoreactive) lymphocytes into the central nervous system (CNS) via α4β1 integrin and into the gut via α4β7 integrin. Natalizumab is approved for the treatment of highly active RRMS, but has also been used in IBD trials, in which the antibody has proved efficient in the induction and maintenance of clinical remission in CD.62,63 However, due to the risk of progressive multifocal leukoencephalopathy (PML) (as a consequence of a reactivation of JC virus), natalizumab is not used in IBD patients.64 Instead, the humanized monoclonal antibody vedolizumab, which only targets the α4β7 integrin–MAdCAM-1 interaction in the intestinal mucosa thereby avoiding the risk of developing PML,65 has proved to be the safer therapeutic option for patients with IBD. As the effect of vedolizumab is limited to the intestine, it is not an option for patients with RRMS.66 Moreover, there have been several cases of MS patients who developed RA or experienced an onset or exacerbation of PsO on treatment with natalizumab.67–70 Natalizumab has been shown to alter the composition of lymphocyte subpopulations in the peripheral blood, with especially CD8+ T and CD19+ B cells being increased.71 These changes – in addition to the hampered migration of leukocytes across the blood–brain or blood–gut barrier – could shift the inflammatory response from the CNS towards other tissues (e.g. the skin or joints), thus triggering the onset of PsO or RA. Thus, natalizumab may be an elegant option for patients with highly active MS and CD, but should be used with caution in patients with comorbid PsO or RA.
TNFα blockers (adalimumab, infliximab, etanercept, golimumab, certolizumab-pegol)
TNFα is a key pro-inflammatory cytokine that induces the production of C-reactive protein, regulates the ability of antigen-presenting cells to activate T cells and induces the production of various pro-inflammatory cytokines, chemokines and cell-adhesion molecules.72 Hence, blocking TNFα with monoclonal antibodies (adalimumab, infliximab, golimumab and certolizumab-pegol) or the soluble TNFα receptor–IgG fusion protein etanercept, has proved to be a very effective therapeutic strategy for several autoimmune conditions, including PsO, RA and IBD73,74 (Table 1). Moreover, TNFα has an even broader range of effects, as tumor necrosis factor alpha receptors (TNFRs) are expressed in almost every cell type.75 However, there are multiple case reports of TNFα blockers promoting or exacerbating demyelinating diseases.6–8 The potential underlying mechanism was unravelled by the discovery of a MS-associated Δ6-TNFR1 genetic variant.76 This gene encodes a soluble protein comprising the extracellular TNFR1 domain (TNF receptor 1), and acts as a decoy receptor for TNFα (similar to etanercept), thus preventing potentially neuroprotective signalling through membrane-bound TNFR1. Furthermore, MS is positively associated with many other TNFα gene variants.77 Therefore, an ongoing anti-TNFα therapy (for PsO, RA or IBD) should be discontinued in patients with coexisting MS.
Tocilizumab and satralizumab
Tocilizumab and satralizumab are monoclonal humanized IL-6 receptor antibodies, which have become a valuable therapeutic asset in several inflammatory conditions, including RA,78 giant cell arteritis (GCA) and aquaropin-4-positive NMOSD.79 Although there are a few case reports describing a presumably secondary CNS autoimmunity on tocilizumab treatment, it has to be taken into account that one of these patients was treated with TNFα blockers prior to tocilizumab and two patients had an already established MS and relapsed during the treatment.80–82 On the other hand, there are also reports of a successful MS therapy with tocilizumab;81,83 there is further experimental evidence showing that blockage of IL-6R can be protective of EAE.84 However, considering the lack of clinical data, we cannot recommend the general use of tocilizumab in patients with MS and RA.
Secukinumab
Secukinumab is a fully human monoclonal IgG1κ IL-17A antibody, which has demonstrated efficacy in phase II and III trials of PsO, psoriasis arthritis and RA.85–87 The IL23/Th17 axis plays a major pathophysiological role in many autoimmune disorders and has particularly been linked to disease severity in PsO and MS.2,3 In fact, Th17 cells were initially described in the EAE model.88,89 Secukinumab has demonstrated promising trends in a phase II trial with RRMS patients;4 although the study’s primary endpoint was not reached. Moreover, there are several case reports that describe a successful treatment of comorbid PsO/MS with secukinumab.90–94 All patients described had a relapsing–remitting disease course and remained clinically and radiologically stable during their respective follow-up periods. Thus, more advanced monoclonal IL-17 antibodies with improved pharmacodynamics, such as brodalumab (anti-17A) or ixekizumab (anti-17A) may also be considered for application in MS. Hence, secukinumab may be one of the best therapeutic options for patients with PsO and coexisting RRMS.
Ustekinumab
Ustekinumab is a fully human monoclonal antibody, targeting the p40 epitope of IL-12 and IL-23. Therefore, ustekinumab blocks the differentiation of naive CD4+ lymphocytes into Th1 and Th17 cells and has shown clinical efficacy in phase III studies for PsO and IBD.95–97 In MS animal models, ustekinumab has proved effective98 and IL-23R−/− mice are even protected from EAE.99,100 Yet ustekinumab failed to transfer those results to a clinical phase II trial.101 This was further corroborated in an EAE model with mice lacking the p40-ustekinumab target epitope.102 However, the patients included in the ustekinumab study had a chronic disease course with long disease durations with low relapse activity. Furthermore, it is not clear whether the antibody can cross the blood–brain barrier effectively. Nevertheless, there are two case reports of patients with MS and comorbid PsO successfully treated with ustekinumab.103 Interestingly, these patients also had a chronic disease course, but did not experience disease progression over a follow-up period of 3–4 years. Taken together, ustekinumab and potentially also other IL-23 antibodies (risakinzumab, tildrakizumab and guselkumab) may have beneficial effects in early MS, but further research needs to be conducted.
Oral/injectable MS therapeutics
Fumaric acid esters
The anti-inflammatory effect of fumarates on psoriatic lesions was initially discovered in the late 1950s104 and it took almost half a century until dimethyl fumarate (DMF) was finally introduced as a potent MS therapeutic.105 DMF exerts its anti-inflammatory effects through activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) transcriptional pathway and the interaction with hydroxycarboxylic acid receptor 2 (HCAR2),106,107 which fosters the upregulation of antioxidant and cytoprotective response genes108 and shifts T cells from a Th1/Th17 profile towards a Th2 phenotype. This was further corroborated by experimental results in murine PsO and EAE models as well as in humans.109 Fumarates are approved for the treatment of mild to moderate RRMS, moderate to severe PsO110–112 (although both being approved in distinct disease-specific drug formulations) and have shown promising results in a model of experimental colitis.113 Furthermore, DMF is in phase II clinical testing for the treatment of RA.114 As DMF is approved for both RRMS and PsO, it is the obvious choice for patients with this particular comorbidity, as exemplified by case reports of patients who experienced remission of PsO flares and additionally remained MS relapse-free on a treatment with DMF.115,116 However, especially older patients may be at risk of developing PML.
Interferons
Interferons (IFNs) are long established DMTs for the treatment of mild to moderately active MS. Similar to results in EAE models, IFNs have shown efficacy in murine models of experimental arthritis,117,118 yet – unlike in MS – these results were either not replicated in RA patients119 or showed conflicting results.120–122 The same holds true for IBDs and PsO.123–128 Exacerbations of autoimmune conditions during an IFN treatment are actually not uncommon and have also been shown for NMOSD.129,130 In PsO, this may be due to a direct involvement of IFNs in PsO pathophysiology: type 1 interferons (IFNα and IFNβ) are key cytokines induced during skin damage; psoriatic lesions can be triggered by skin injury, also known as ‘Köbners phenomenon’.131 Furthermore, the type 1 IFN pathway is necessary for the development of psoriasis-like inflammatory diseases in mice.132,133 Ultraviolet (UV) light therapy, on the other hand, ameliorates PsO flares, as it triggers ubiquitination and downregulation of the type 1 interferon receptor IFNAR1 and thus IFN signalling.134 Yet despite these negative associations, an established IFN therapy in MS patients should not necessarily be discontinued when a second autoimmune condition is diagnosed.
S1PR modulators (fingolimod, siponimod, ozanimod)
S1PR modulators cause an internalization and degradation of S1PRs, thus trapping lymphocytes in secondary lymphoid organs (e.g. lymph nodes).135 This reduces the number of circulating lymphocytes and ultimately precludes their invasion into the CNS. In 2010, the non-selective S1PR modulator fingolimod was the first orally administered drug approved for the treatment of highly active RRMS.136 Since then, siponimod and now also ozanimod have been approved as DMTs for patients with secondary progressive multiple sclerosis (SPMS) and RRMS, respectively. While S1PR modulators have shown promising results in MS, they also have the potential to be useful DMTs in IBD, RA or PsO, as recently demonstrated in various animal models.137–142 While there are no clinical data on S1PR modulators in RA, and two studies regarding PsO,143,144 the TOUCHSTONE (NCT01647516) and STEPSTONE (NCT02531113), as well as the ‘True North’ trials demonstrated that ozanimod is safe, well tolerated and effective in patients with moderate to severe UC.5,145 Taken together, especially ozanimod could be an elegant monotherapy option for patients with highly active MS and moderate to severe IBD.
Teriflunomide and leflunomide
Teriflunomide and leflunomide reversibly inhibit the mitochondrial enzyme dihydroorotate dehydrogenase (DHODH), causing a decreased de novo pyrimidine synthesis, thus diminishing T- and B cell activation and proliferation.146 Teriflunomide is approved for the treatment of mild to moderate RRMS, whereas its pro-drug leflunomide is in clinical use for the treatment of RA and psoriatic arthritis.147,148 Gastrointestinal symptoms are among the most common unwanted side effects of teriflunomide, but these events are usually mild and self-limiting. However, there are a few cases of severe (lymphocytic) colitis with teriflunomide treatment in patients with RRMS,149,150 and teriflunomide has also been associated with the onset of PsO.151,152 However, as these are single case reports, a discontinuation of teriflunomide in patients with comorbid PsO and IBD should be carefully considered, while in patients with mild to moderate RRMS and coexisting RA, teriflunomide remains a valid therapeutic option.
Discussion
In the following, we suggest three pragmatic treatment approaches for patients with MS and coexisting autoimmune disorders (Figures 1–3). After exclusion of contraindicated drugs, the therapeutics should be chosen according to the individual patient’s disease activity. Table 2 shows the safety rating. Table 1 shows the FDA approval status of the respective immunosuppressants. Overall, TNFα blockers are effectively utilized in PsO, RA and IBD, yet have doubtlessly been shown to exacerbate or induce demyelination. Thus, TNFα blockers are the only real absolute contraindication for all patients with coexisting MS (Figures 1–3; Table 2).
Figure 1.
Flow chart showing the decision-making process for patients with MS and PsO. Descriptions are given in the main text.
DMF, dimethyl fumarate; JAK, Janus kinase; MS, multiple sclerosis; MTX, methotrexate; PsO, psoriasis; S1PR, sphingosine-1-phosphate receptor modulator.
Figure 2.
Flow chart showing the decision-making process for patients with MS and RA. Descriptions are given in the main text.
DMF, dimethyl fumarate; HCQ, hydroxychloroquine; JAK, Janus kinase; MS, multiple sclerosis; MTX, methotrexate; RA, rheumatoid arthritis; S1PR, sphingosine-1-phosphate receptor modulator.
Figure 3.
Flow chart showing the decision-making process for patients with MS and IBD. Descriptions are given in the main text.
AZA, azathioprine; CYA, cyclosporine A; IBD, inflammatory bowel disease; JAK, Janus kinase; MS, multiple sclerosis; MTX, methotrexate.
Table 2.
Efficacy and safety rating for each disease-modifying therapy with regard to use in the main autoimmune disorders discussed here.
| MS | IBD | PsO | RA | |
|---|---|---|---|---|
| General immunosuppressants | ||||
| MTX | + | + | + | ++ |
| AZA | + | ++ | + | + |
| MMF | + | + | + | + |
| 5-ASA | (+/−) | ++ | (−) | + |
| HCQ | (+) | (+) | – | + |
| CYA/TAC | + | ++ | ++ | ++ |
| Oral/injectable MS therapeutics | ||||
| IFN | ++ | (−) | (−) | (−) |
| TER/LEF | ++ | (−) | + | ++ |
| DMF | ++ | (+) | ++ | (+) |
| S1PR | +++ | ++ | (+) | (+) |
| Monoclonal antibodies | ||||
| TOC | (+/−) | (+) | (−) | + |
| USTE | (+) | ++ | +++ | 0 |
| SEC | + | +/− | +++ | (+) |
| Anti-TNFα | – – – | +++ | +++ | +++ |
| NATA | +++ | ++ | (−) | (−) |
| ALEM | (+++) | x | (+) | (+++) |
| Anti-CD20 | +++ | (−) | (−) | +++ |
| Others | ||||
| JAK | (+) | ++ | ++ | ++ |
| Abatacept | 0 | 0 | + | ++ |
| Anakinra | x | (+) | + | ++ |
Color gradient ranges from absolutely contraindicated (dark red) to highly effective (dark green). – – –, absolutely contraindicated; (−), possibly harmful; (+), possibly effective; (+/−), mixed reports; (+++), highly effective; but restricted label due to safety concerns; –, harmful; +, effective; ++, very effective; +++, highly effective; 0, not effective.
5-ASA, 5-aminosalicylic acid; ALEM, alemtuzumab; Anti-CD20, CD20-antibodies; Anti-TNFα, TNFα antibodies; AZA, azathioprine; CYA/TAC, cyclosporine/tacrolimus; DMF, dimethylfumarate; HCQ, hydroxchloroquine; IBD, inflammatory bowel disease; IFN, interferon; JAK, Janus kinase inhibitor; MMF, mycophenolate mofetil; MS, multiple sclerosis; MTX, methotrexate; NATA, natalizumab; PsO, psoriasis; RA, rheumatoid arthritis; S1PR, sphingosin-1-phosphate receptor modulator; SEC, secukinumab; TER/LEF, teriflunomide/leflunomide; TOC, tocilizumab; USTE, ustekinumab; x, no data.
MS plus PsO (Figure 1)
As a chronic inflammatory skin condition, PsO can be treated not only systemically, but depending on disease severity and the existence of extracutaneous manifestations (e.g. psoriasis–arthritis), topical DMTs are also available. Those therapeutics can be combined with every DMT approved for MS.
Fumarates have proved effective in both RRMS and PsO, thus patients with a moderate disease course could be set on a monotherapy with DMF or alternatively a combination of DMF and MTX or CYA. Patients with highly active RRMS and more severe psoriasis could benefit from a monotherapy with secukinumab or a combination of secukinumab and DMF or MTX. As secukinumab has already been tested in a clinical phase II trial and there are several case reports of its successful application in RRMS patients,90–94 the anti-IL17 antibody has – in comparison with other DMTs – the highest level of clinical evidence. However, it needs to be taken into account that the trial by Havrdova et al.4 failed to reach its primary endpoint [the cumulative number of combined unique active lesions (CUALs) observed in magnetic resonance imaging (MRI) scans within 2 years], while still showing a significant reduction in cumulative new gadolinium-enhancing T1 lesions (secondary endpoint). Hence, larger, better powered trials are still needed to evaluate further the benefit of IL-17 targeting in RRMS patients (e.g. brodalumab, ixekizumab).
S1PR modulators have shown promising results in various PsO mouse models, demonstrating strong anti-proliferative and anti-inflammatory effects, significantly reducing the animals’ disease burden.137,138 This has been further corroborated in clinical studies with PsO patients, which demonstrated a significant reduction of PsO plaque burden on treatment with the S1PR modulator ponesimod;143,144 interestingly, ponesimod is about to be approved for its application in RRMS.153 Thus, in addition to anti-IL17 antibodies, S1PR modulators may be another good alternative for patients with highly active RRMS and coexisting PsO. Patients with concomitant active SPMS and PsO may be treated with siponimod, yet there are no clinical data to support this assertion. Certain subgroups of RRMS patients with comorbid PsO may also profit from a therapy with alemtuzumab, yet there is only one single case report to support this approach.44
There have been several case studies describing an onset or exacerbation of PsO in MS patients treated with CD20 antibodies, teriflunomide, natalizumab and IFNs. However, these studies are mostly based on single case reports, whereas the vast majority of patients did not exhibit comparable unwanted side effects. Thus, the application of those DMTs should be carefully evaluated in patients with MS and coexisting PsO, yet they should not be generally excluded from the therapeutic arsenal, especially when patients have remained relapse free with these substances.
RA plus MS (Figure 2)
The DHODH inhibitors teriflunomide and leflunomide are approved for moderate RRMS and RA, respectively. Even though leflunomide is the pro-drug of teriflunomide and there is vast clinical experience with both DMTs, slight differences between the two substances or the respective patient populations (RA/MS) cannot be completely excluded. However, as DHODH inhibitors are already in clinical application for both conditions, teriflunomide is the obvious choice for patients with moderate RRMS and comorbid RA. Although MTX is not commonly used in patients with RRMS, the DHFR inhibitor has a rather favorable therapeutic profile and is the most frequently used DMT in RA. Thus, MTX could also be utilized as a second line crossover DMT. The same holds true for AZA.
Patients with highly active RRMS and RA have both shown responsiveness to anti-CD20 antibodies (rituximab/ocrelizumab). In RA, rituximab is readily combined with other DMARDs (leflunomide or MTX),15 which could additionally prohibit MS progression, but may also be a source of unwanted side effects and should therefore be carefully evaluated.
Patients with PPMS and RA could profit from ocrelizumab therapy, yet ocrelizumab is only approved for the application in MS. HCQ, which is now only rarely used in RA therapy, may potentially become another option for PPMS in the future, but so far there are only experimental data available. The same is true for S1PR modulators, which have shown promising results in experimental arthritis models, but have not yet been tested for their clinical application in RA. In the early to mid-1990s, alemtuzumab was successfully applied in certain cohorts of therapy-refractory RA patients,42 yet its use has not prevailed due to its unfavorable safety profile and prolonged lymphopenia. However, as mentioned for PsO, certain subgroups of RRMS patients with comorbid RA might profit from a therapy with alemtuzumab.
The CTLA4 fusion protein abatacept and the recombinant IL-1 receptor antagonist anakinra are both approved for RA and have also been tested in patients with RRMS, yet have either failed to show beneficial effects on disease progression,154 or the trials are still ongoing (anakinra: NCT04025554).
There are several case reports describing an onset or exacerbation of RA in MS patients treated with natalizumab and interferons.70,120–122 However, as discussed for PsO, these are single case reports and a general discontinuation of an already established MS therapy – after RA onset – should be carefully evaluated.
IBDs plus MS (Figure 3)
While mild cases of IBD can be treated with local (rectal) glucocorticoids (e.g. budesonide) and/or salicylates (5-ASA), severe, refractory cases may need surgical attention. As mentioned for PsO, these topically administered substances should be continued throughout the MS therapy. However, the standard relapse prophylactic for IBDs – oral 5-ASA – did not show a significant effect on MS disease progression and is thus not suitable for a combinatory treatment.
While AZA is no longer commonly used as a DMT in MS, the thiopurine is readily utilized as a monotherapy to maintain remission in IBD and is therefore the evident option for a combinatory therapy. If AZA is not well tolerated, MTX remains a possible secondary option.155 The calcineurine inhibitor CYA is approved as a remission-inducing agent in severe, steroid refractory UC.16,17 Yet, because of the side effects of CYA, it is not generally used as a long-term therapeutic; remission maintenance is usually achieved instead with thiopurines or vedolizumab. Hence, CYA is not an optimal DMT for patients with coexisting MS and IBD, but may be carefully considered in certain cases.
Natalizumab has proved effective in the induction and maintenance of remission in IBDs. Hence, a natalizumab monotherapy is a valid therapeutic option for patients with highly active RRMS and IBD. Alternatively, ozanimod has recently been approved for both RRMS and moderate to severe IBD, thus especially patients with high documented JC virus titers could benefit from a treatment with the S1PR modulator.
Due to frequently reported gastrointestinal side effects and recent reports of refractory lymphocytic colitis under teriflunomide,149,150 the DHOD inhibitor may not be the best therapeutic choice for patients with RRMS and comorbid IBD. The same is true for CD20 antibodies, which have also been associated with the onset of colitis. This is further corroborated by several animal models, demonstrating that B-cell depletion can deregulate the gastrointestinal immune system by increasing inflammatory cytokines in both blood serum and intestinal tissue.156,157 Nevertheless, those cases are also reports of rare instances and should not be over-interpreted.
The immunomodulatory drug glatiramer acetate (GLAT), is commonly used in patients with moderate RRMS, but has – from a pathophysiology perspective – no clear application in other autoimmune disorders. Nevertheless, there are a few studies in IBD animal models that demonstrate a therapeutic effect of GLAT158,159 via the induction of regulatory T cells.160 Thus, GLAT may potentially have a broader application outside of MS, but so far there are only experimental data available.
Future perspectives
As therapeutic strategies for cancer and autoimmune diseases of different organs target similar pathways and thus show some parallels, novel approaches in one of these fields often emerge from neighboring disciplines and benefit from experiences made there. The latter, together with distinct contrasting effects of treatments in different diseases, provides valuable insight for overlapping clinical conditions with more than one disease. Targeting specific intracellular signalling cascades or tyrosine kinases with small molecule inhibitors has become a general therapeutic strategy in medical oncology. Thus, as shown for former chemotherapeutics – such as MTX or AZA – this trend has now also spread to autoimmune disorders, as demonstrated by Janus kinase inhibitors.
Janus kinases (JAKs) are cytoplasmic protein tyrosine kinases that mediate the induction of STAT proteins, which then activate the transcription of various pro-inflammatory cytokines (e.g. INFγ or TNFα). JAK inhibitors, such as tofacitinib, ruxolitinib or baricitinib, have shown efficacy in the treatment of several inflammatory conditions and are approved for the treatment of PsO, RA and UC.161–163 In general, these small molecule inhibitors are less effective than biologicals, yet have proved to be advantageous because of their oral/topical administration and overall cost-effectiveness.164 Although there have been conflicting results for JAK inhibitors in EAE models and MS patients,165,166 the JAK/STAT pathway is critically involved in MS pathophysiology,167 thus JAK inhibitors may also become available for MS in the future.
In the past decades, immunotherapy has become more and more precise, targeting specific cytokines (e.g. IL-17) or cell types (e.g. B cells). According to numerous experimental animal models, MS was primarily considered a T-cell-mediated disease. However, there is an increasing amount of evidence for the importance of B cells and humoral immunity in the genesis of demyelinating lesions, making especially the BAFF/APRIL system a possible therapeutic target.168 B cell-activating factor (BAFF) and a proliferation-inducing ligand (APRIL) are inflammatory cytokines, which are primarily involved in B cell survival, maturation and activity.169 Yet, thus far targeting BAFF and/or APRIL with monoclonal antibodies or a soluble decoy TACI-Fc fusion protein did not bring the desired results in MS patients.170,171 One possible explanation is that the soluble decoy TACI-Fc fusion protein atacicept only has a B cell subset-specific effect and might have disturbed the fine-tuned balance of conventional and regulatory B cells, resulting in excessive T cell responses. Furthermore, the BAFF antibody tabalumab, as well as atacicept, spares memory B cells, thus T cell activation may still be preserved.170 Similar results and conclusions have been obtained for the application of atacicept and tabalumab in RA.172 Nevertheless, in spite of these negative results, CD20 antibodies have demonstrated the efficacy of B cell targeting therapies, thus we may see the emergence of new B cell-associated therapeutics for MS and RA in the future. In this regard, the Burton tyrosine kinase (BTK) is critically involved in B cell signalling and small molecule BTK inhibitors (e.g. tolebrutinib, evobrutinib) have shown promising results in preclinical MS and also RA models173 as well as in clinical trials.174 Moreover, there are several ongoing phase III trials on BTK inhibitors in MS. Thus, BTK inhibitors may soon become readily used substances in MS, RA and potentially also other inflammatory conditions.
Although it is not uncommon for MS patients to present with comorbid autoimmune disorders such as PsO, RA, and IBD, the likelihood of recruiting sufficient numbers of these patients for large-scale clinical trials in the foreseeable future is low. This emphasizes the importance of case reports and review articles, until the growing arsenal of new immunosuppressants is able to provide more straightforward treatment approaches for patients with MS and coexisting autoimmune disorders.
Acknowledgments
The authors thank Cheryl Ernest for proofreading and editing the manuscript.
Footnotes
Conflict of interest statement: TB has no competing interests to declare. TR received travel grants and financial research support from Genzyme and Novartis and received honoraria for lecturing from Roche, Merck, Genzyme, Biogen and Teva. SGM received honoraria for lecturing and travel expenses for attending meetings from Almirall, Amicus Therapeutics Germany, Bayer Health Care, Biogen, Celgene, Diamed, Genzyme, MedDay Pharmaceuticals, Merck Serono, Novartis, Novo Nordisk, ONO Pharma, Roche, Sanofi-Aventis, Chugai Pharma, QuintilesIMS and Teva. His research is funded by the German Ministry for Education and Research (BMBF), Deutsche Forschungsgemeinschaft (DFG), Else Kröner Fresenius Foundation, German Academic Exchange Service, Hertie Foundation, Interdisciplinary Center for Clinical Studies (IZKF) Muenster, German Foundation Neurology and by Almirall, Amicus Therapeutics Germany, Biogen, Diamed, Fresenius Medical Care, Genzyme, Merck Serono, Novartis, ONO Pharma, Roche and Teva. FZ has recently received research grants and/or consultation funds from DFG, BMBF, PMSA, MPG, Genzyme, Merck Serono, Roche, Novartis, Sanofi-Aventis, Celgene, ONO and Octapharma. SB has received honoraria and compensation for travel from Biogen Idec, Merck Serono, Novartis, Sanofi-Genzyme and Roche.
Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was supported by the German Research Council (DFG, CRC-TR-128 to FZ, SGM and SB) and Hertie-Stiftung (SB).
ORCID iD: Stefan Bittner
https://orcid.org/0000-0003-2179-3655
Contributor Information
Tobias Brummer, Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
Tobias Ruck, Department of Neurology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.
Sven G. Meuth, Department of Neurology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
Frauke Zipp, Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
Stefan Bittner, Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, Rhineland-Palatinate, Mainz 55131, Germany.
References
- 1.Gutierrez-Arcelus M, Rich SS, Raychaudhuri S. Autoimmune diseases – connecting risk alleles with molecular traits of the immune system. Nat Rev Genet 2016; 17: 160–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Mahil SK, Capon F, Barker JN. Update on psoriasis immunopathogenesis and targeted immunotherapy. Semin Immunopathol 2016; 38: 11–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Hiltensperger M, Korn T. The interleukin (IL)-23/T helper (Th)17 axis in experimental autoimmune encephalomyelitis and multiple sclerosis. Cold Spring Harb Perspect Med 2018; 8: a029637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Havrdova E, Belova A, Goloborodko A, et al. Activity of secukinumab, an anti-IL-17A antibody, on brain lesions in RRMS: results from a randomized, proof-of-concept study. J Neurol 2016; 263: 1287–1295. [DOI] [PubMed] [Google Scholar]
- 5.Sandborn WJ, Feagan BG, Wolf DC, et al. Ozanimod induction and maintenance treatment for ulcerative colitis. N Engl J Med 2016; 374: 1754–1762. [DOI] [PubMed] [Google Scholar]
- 6.Caminero A, Comabella M, Montalban X. Tumor necrosis factor alpha (TNF-α), anti-TNF-α and demyelination revisited: an ongoing story. J Neuroimmunol 2011; 234: 1–6. [DOI] [PubMed] [Google Scholar]
- 7.Kopp TI, Delcoigne B, Arkema EV, et al. Risk of neuroinflammatory events in arthritis patients treated with tumour necrosis factor alpha inhibitors: a collaborative population-based cohort study from Denmark and Sweden. Ann Rheum Dis 2020; 79: 566–572. [DOI] [PubMed] [Google Scholar]
- 8.Toussirot E, Pertuiset E, Martin A, et al. Association of rheumatoid arthritis with multiple sclerosis: report of 14 cases and discussion of its significance. J Rheumatol 2006; 33: 1027–1028. [PubMed] [Google Scholar]
- 9.Willkens RF, Urowitz MB, Stablein DM, et al. Comparison of azathioprine, methotrexate, and the combination of both in the treatment of rheumatoid arthritis. A controlled clinical trial. Arthritis Rheum 1992; 35: 849–856. [DOI] [PubMed] [Google Scholar]
- 10.Mandler RN, Ahmed W, Dencoff JE. Devic’s neuromyelitis optica: a prospective study of seven patients treated with prednisone and azathioprine. Neurology 1998; 51: 1219–1220. [DOI] [PubMed] [Google Scholar]
- 11.Bichuetti DB, Lobato de, Oliveira EM, Oliveira DM, et al. Neuromyelitis optica treatment: analysis of 36 patients. Arch Neurol 2010; 67: 1131–1136. [DOI] [PubMed] [Google Scholar]
- 12.Massacesi L, Tramacere I, Amoroso S, et al. Azathioprine versus beta interferons for relapsing–remitting multiple sclerosis: a multicentre randomized non-inferiority trial. PLoS One 2014; 9: e113371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Messori A, Fadda V, Maratea D, et al. Indirect meta-analytical comparison of azathioprine and of beta interferon effectiveness in all forms of multiple sclerosis pooled together. J Neurol Sci 2014; 347: 408–410. [DOI] [PubMed] [Google Scholar]
- 14.Confavreux C, Saddier P, Grimaud J, et al. Risk of cancer from azathioprine therapy in multiple sclerosis: a case–control study. Neurology 1996; 46: 1607–1612. [DOI] [PubMed] [Google Scholar]
- 15.Meier FM, Frerix M, Hermann W, et al. Current immunotherapy in rheumatoid arthritis. Immunotherapy 2013; 5: 955–974. [DOI] [PubMed] [Google Scholar]
- 16.Narula N, Marshall JK, Colombel JF, et al. Systematic review and meta-analysis: infliximab or cyclosporine as rescue therapy in patients with severe ulcerative colitis refractory to steroids. Am J Gastroenterol 2016; 111: 477–491. [DOI] [PubMed] [Google Scholar]
- 17.Wu B, Tong J, Ran Z. Tacrolimus therapy in steroid-refractory ulcerative colitis: a review. Inflamm Bowel Dis 2020; 26: 24–32. [DOI] [PubMed] [Google Scholar]
- 18.Nast A, Smith C, Spuls PI, et al. EuroGuiDerm guideline on the systemic treatment of Psoriasis vulgaris – part 1: treatment and monitoring recommendations. J Eur Acad Dermatol Venereol 2020; 34: 2461–2498. [DOI] [PubMed] [Google Scholar]
- 19.Zhao GJ, Li DK, Wolinsky JS, et al. Clinical and magnetic resonance imaging changes correlate in a clinical trial monitoring cyclosporine therapy for multiple sclerosis. The MS Study Group. J Neuroimaging 1997; 7: 1–7. [DOI] [PubMed] [Google Scholar]
- 20.Currier RD, Haerer AF, Meydrech EF. Low dose oral methotrexate treatment of multiple sclerosis: a pilot study. J Neurol Neurosurg Psychiatry 1993; 56: 1217–1218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Jacques F, Gaboury I, Christie S, et al. Combination therapy of interferon Beta-1b and tacrolimus: a pilot safety study. Mult Scler Int 2012; 2012: 935921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Suarez-Almazor ME, Belseck E, Shea B, et al. Sulfasalazine for rheumatoid arthritis. Cochrane Database Syst Rev 2000: CD000958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Koch MW, Zabad R, Giuliani F, et al. Hydroxychloroquine reduces microglial activity and attenuates experimental autoimmune encephalomyelitis. J Neurol Sci 2015; 358: 131–137. [DOI] [PubMed] [Google Scholar]
- 24.Faissner S, Mahjoub Y, Mishra M, et al. Unexpected additive effects of minocycline and hydroxychloroquine in models of multiple sclerosis: prospective combination treatment for progressive disease? Mult Scler 2018; 24: 1543–1556. [DOI] [PubMed] [Google Scholar]
- 25.Brown D, Moezzi D, Dong Y, et al. Combination of hydroxychloroquine and indapamide attenuates neurodegeneration in models relevant to multiple sclerosis. Neurotherapeutics 2021; 18: 387–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Ashtari F, Savoj MR. Effects of low dose methotrexate on relapsing–remitting multiple sclerosis in comparison to Interferon β-1α: a randomized controlled trial. J Res Med Sci 2011; 16: 457–462. [PMC free article] [PubMed] [Google Scholar]
- 27.Goodkin DE, Rudick RA, VanderBrug Medendorp S, et al. Low-dose oral methotrexate in chronic progressive multiple sclerosis: analyses of serial MRIs. Neurology 1996; 47: 1153–1157. [DOI] [PubMed] [Google Scholar]
- 28.Palaniappan S, Ford AC, Greer D, et al. Mycophenolate mofetil therapy for refractory inflammatory bowel disease. Inflamm Bowel Dis 2007; 13: 1488–1492. [DOI] [PubMed] [Google Scholar]
- 29.Marinari R, Fleischmajer R, Schragger AH, et al. Mycophenolic acid in the treatment of psoriasis: long-term administration. Arch Dermatol 1977; 113: 930–932. [PubMed] [Google Scholar]
- 30.Goldblum R. Therapy of rheumatoid arthritis with mycophenolate mofetil. Clin Exp Rheumatol 1993; 11 (Suppl. 8): S117–S119. [PubMed] [Google Scholar]
- 31.Michel L, Vukusic S, De Seze J, et al. Mycophenolate mofetil in multiple sclerosis: a multicentre retrospective study on 344 patients. J Neurol Neurosurg Psychiatry 2014; 85: 279–283. [DOI] [PubMed] [Google Scholar]
- 32.Fakih R, Matiello M, Chitnis T, et al. Efficacy and safety of mycophenolate mofetil in progressive multiple sclerosis patients. J Neurol 2018; 265: 2688–2694. [DOI] [PubMed] [Google Scholar]
- 33.Ko CW, Singh S, Feuerstein JD, et al. AGA clinical practice guidelines on the management of mild-to-moderate ulcerative colitis. Gastroenterology 2019; 156: 748–764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Rains CP, Noble S, Faulds D. Sulfasalazine. A review of its pharmacological properties and therapeutic efficacy in the treatment of rheumatoid arthritis. Drugs 1995; 50: 137–156. [DOI] [PubMed] [Google Scholar]
- 35.Kim S, Lee YI, Chang KY, et al. Promotion of remyelination by sulfasalazine in a transgenic Zebrafish model of demyelination. Mol Cells 2015; 38: 1013–1021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Prosiegel M, Neu I, Vogl S, et al. Suppression of experimental autoimmune encephalomyelitis by sulfasalazine. Acta Neurol Scand 1990; 81: 237–238. [DOI] [PubMed] [Google Scholar]
- 37.Correale J, Olsson T, Bjork J, et al. Sulfasalazine aggravates experimental autoimmune encephalomyelitis and causes an increase in the number of autoreactive T cells. J Neuroimmunol 1991; 34: 109–120. [DOI] [PubMed] [Google Scholar]
- 38.Uitdehaag BM, Polman CH, de Groot CJ, et al. Failure of sulfasalazine to influence experimental autoimmune encephalomyelitis. Acta Neurol Scand 1991; 84: 173–174. [DOI] [PubMed] [Google Scholar]
- 39.Noseworthy JH, O’Brien P, Erickson BJ, et al. The Mayo Clinic–Canadian cooperative trial of sulfasalazine in active multiple sclerosis. Neurology 1998; 51: 1342–1352. [DOI] [PubMed] [Google Scholar]
- 40.Gold R, Kappos L, Becker T. Development of multiple sclerosis in patient on long-term sulfasalazine. Lancet 1990; 335: 409–410. [DOI] [PubMed] [Google Scholar]
- 41.Investigators CT, Coles AJ, Compston DA, et al. Alemtuzumab vs. interferon beta-1a in early multiple sclerosis. N Engl J Med 2008; 359: 1786–1801. [DOI] [PubMed] [Google Scholar]
- 42.Weinblatt ME, Maddison PJ, Bulpitt KJ, et al. CAMPATH-1H, a humanized monoclonal antibody, in refractory rheumatoid arthritis. An intravenous dose-escalation study. Arthritis Rheum 1995; 38: 1589–1594. [DOI] [PubMed] [Google Scholar]
- 43.Cooles FA, Anderson AE, Drayton T, et al. Immune reconstitution 20 years after treatment with alemtuzumab in a rheumatoid arthritis cohort: implications for lymphocyte depleting therapies. Arthritis Res Ther 2016; 18: 302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Aslam A, Marsland AM, Rog D, et al. Alemtuzumab and chronic plaque psoriasis. Br J Dermatol 2013; 169: 184–186. [DOI] [PubMed] [Google Scholar]
- 45.Zhu WM, Li Y, Yu C, et al. Antimouse CD52 monoclonal antibody inhibits established spontaneous colitis in IL-10-deficient mice. Inflamm Bowel Dis 2011; 17: E72–E73. [DOI] [PubMed] [Google Scholar]
- 46.Salles G, Barrett M, Foa R, et al. Rituximab in B-cell hematologic malignancies: a review of 20 years of clinical experience. Adv Ther 2017; 34: 2232–2273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Borisow N, Mori M, Kuwabara S, et al. Diagnosis and treatment of NMO spectrum disorder and MOG-encephalomyelitis. Front Neurol 2018; 9: 888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Arneth BM. Impact of B cells to the pathophysiology of multiple sclerosis. J Neuroinflammation 2019; 16: 128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Hauser SL, Waubant E, Arnold DL, et al. B-cell depletion with rituximab in relapsing–remitting multiple sclerosis. N Engl J Med 2008; 358: 676–688. [DOI] [PubMed] [Google Scholar]
- 50.Chisari CG, Sgarlata E, Arena S, et al. Rituximab for the treatment of multiple sclerosis: a review. J Neurol. Epub ahead of print 8January2021. DOI: 10.1007/s00415-020-10362-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Montalban X, Belachew S, Wolinsky JS. Ocrelizumab in primary progressive and relapsing multiple sclerosis. N Engl J Med 2017; 376: 1694. [DOI] [PubMed] [Google Scholar]
- 52.Hauser SL, Bar-Or A, Comi G, et al. Ocrelizumab versus interferon beta-1a in relapsing multiple sclerosis. N Engl J Med 2017; 376: 221–234. [DOI] [PubMed] [Google Scholar]
- 53.Abushouk AI, Ahmed H, Ismail A, et al. Safety and efficacy of ocrelizumab in rheumatoid arthritis patients with an inadequate response to methotrexate or tumor necrosis factor inhibitors: a systematic review and meta-analysis. Rheumatol Int 2017; 37: 1053–1064. [DOI] [PubMed] [Google Scholar]
- 54.Kristjansson VB, Lund SH, Grondal G, et al. Increased risk of inflammatory bowel disease among patients treated with rituximab in Iceland from 2001 to 2018. Scand J Gastroenterol 2021; 56: 46–52. [DOI] [PubMed] [Google Scholar]
- 55.Venables ZC, Swart SS, Soon CS. Palmoplantar pustulosis secondary to rituximab: a case report and literature review. Clin Exp Dermatol 2015; 40: 451–452. [DOI] [PubMed] [Google Scholar]
- 56.Alahmari HS, Alhowaish NY, Omair MA. Rituximab-induced psoriasis in a patient with granulomatosis with polyangitis treated with adalimumab. Case Rep Rheumatol 2019; 2019: 5450863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Darwin E, Romanelli P, Lev-Tov H. Ocrelizumab-induced psoriasiform dermatitis in a patient with multiple sclerosis. Dermatol Online J 2018; 24: 13030/qt220859qb. [PubMed] [Google Scholar]
- 58.Sunjaya DB, Taborda C, Obeng R, et al. First case of refractory colitis caused by ocrelizumab. Inflamm Bowel Dis 2020; 26: e49. [DOI] [PubMed] [Google Scholar]
- 59.Rosser EC, Oleinika K, Tonon S, et al. Regulatory B cells are induced by gut microbiota-driven interleukin-1beta and interleukin-6 production. Nat Med 2014; 20: 1334–1339. [DOI] [PubMed] [Google Scholar]
- 60.Wang L, Ray A, Jiang X, et al. T regulatory cells and B cells cooperate to form a regulatory loop that maintains gut homeostasis and suppresses dextran sulfate sodium-induced colitis. Mucosal Immunol 2015; 8: 1297–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Aira LE, Debes GF. Skin-homing regulatory B cells required for suppression of cutaneous inflammation. J Invest Dermatol. Epub ahead of print 9February2021. DOI: 10.1016/j.jid.2021.01.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Sandborn WJ, Colombel JF, Enns R, et al. Natalizumab induction and maintenance therapy for Crohn’s disease. N Engl J Med 2005; 353: 1912–1925. [DOI] [PubMed] [Google Scholar]
- 63.Targan SR, Feagan BG, Fedorak RN, et al. Natalizumab for the treatment of active Crohn’s disease: results of the ENCORE trial. Gastroenterology 2007; 132: 1672–1683. [DOI] [PubMed] [Google Scholar]
- 64.Bloomgren G, Richman S, Hotermans C, et al. Risk of natalizumab-associated progressive multifocal leukoencephalopathy. N Engl J Med 2012; 366: 1870–1880. [DOI] [PubMed] [Google Scholar]
- 65.Feagan BG, Greenberg GR, Wild G, et al. Treatment of ulcerative colitis with a humanized antibody to the α4β7 integrin. N Engl J Med 2005; 352: 2499–2507. [DOI] [PubMed] [Google Scholar]
- 66.Keeley KA, Rivey MP, Allington DR. Natalizumab for the treatment of multiple sclerosis and Crohn’s disease. Ann Pharmacother 2005; 39: 1833–1843. [DOI] [PubMed] [Google Scholar]
- 67.Vacchiano V, Foschi M, Sabattini L, et al. Arthritic psoriasis during natalizumab treatment: a case report and review of the literature. Neurol Sci 2018; 39: 181–183. [DOI] [PubMed] [Google Scholar]
- 68.Lambrianides S, Kinnis E, Leonidou E, et al. Does natalizumab induce or aggravate psoriasis? A case study and review of the literature. Case Rep Neurol 2018; 10: 286–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Millan-Pascual J, Turpin-Fenoll L, Del Saz-Saucedo P, et al. Psoriasis during natalizumab treatment for multiple sclerosis. J Neurol 2012; 259: 2758–2760. [DOI] [PubMed] [Google Scholar]
- 70.Su E, Novic J, Han MH. Emergence of rheumatoid arthritis following exposure to natalizumab. Mult Scler Relat Disord 2020; 40: 101936. [DOI] [PubMed] [Google Scholar]
- 71.Kaufmann M, Haase R, Proschmann U, et al. Real-world lab data in natalizumab treated multiple sclerosis patients up to 6 years long-term follow up. Front Neurol 2018; 9: 1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Summers deLuca L, Gommerman JL. Fine-tuning of dendritic cell biology by the TNF superfamily. Nat Rev Immunol 2012; 12: 339–351. [DOI] [PubMed] [Google Scholar]
- 73.Hetland ML, Christensen IJ, Tarp U, et al. Direct comparison of treatment responses, remission rates, and drug adherence in patients with rheumatoid arthritis treated with adalimumab, etanercept, or infliximab: results from eight years of surveillance of clinical practice in the nationwide Danish DANBIO registry. Arthritis Rheum 2010; 62: 22–32. [DOI] [PubMed] [Google Scholar]
- 74.Abanmi A, Al Harthi F, Al Agla R, et al. Serum levels of proinflammatory cytokines in psoriasis patients from Saudi Arabia. Int J Dermatol 2005; 44: 82–83. [DOI] [PubMed] [Google Scholar]
- 75.Eissner G, Kolch W, Scheurich P. Ligands working as receptors: reverse signaling by members of the TNF superfamily enhance the plasticity of the immune system. Cytokine Growth Factor Rev 2004; 15: 353–366. [DOI] [PubMed] [Google Scholar]
- 76.Gregory AP, Dendrou CA, Attfield KE, et al. TNF receptor 1 genetic risk mirrors outcome of anti-TNF therapy in multiple sclerosis. Nature 2012; 488: 508–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Caminero A, Comabella M, Montalban X. Role of tumour necrosis factor (TNF)-α and TNFRSF1A R92Q mutation in the pathogenesis of TNF receptor-associated periodic syndrome and multiple sclerosis. Clin Exp Immunol 2011; 166: 338–345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Scott LJ. Tocilizumab: a review in rheumatoid arthritis. Drugs 2017; 77: 1865–1879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Duchow A, Bellmann-Strobl J. Satralizumab in the treatment of neuromyelitis optica spectrum disorder. Neurodegener Dis Manag 2021; 11: 49–59. [DOI] [PubMed] [Google Scholar]
- 80.Beauchemin P, Carruthers R. MS arising during Tocilizumab therapy for rheumatoid arthritis. Mult Scler 2016; 22: 254–256. [DOI] [PubMed] [Google Scholar]
- 81.Sato H, Kobayashi D, Abe A, et al. Tocilizumab treatment safety in rheumatoid arthritis in a patient with multiple sclerosis: a case report. BMC Res Notes 2014; 7: 641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Burmester GR, Feist E, Kellner H, et al. Effectiveness and safety of the interleukin 6-receptor antagonist tocilizumab after 4 and 24 weeks in patients with active rheumatoid arthritis: the first phase IIIb real-life study (TAMARA). Ann Rheum Dis 2011; 70: 755–759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Hoshino H, Shirai Y, Konishi H, et al. Efficacy of tocilizumab for fulminant multiple sclerosis with a tumefactive cervical lesion: a 12-year-old boy. Mult Scler Relat Disord 2020; 37: 101460. [DOI] [PubMed] [Google Scholar]
- 84.Serada S, Fujimoto M, Mihara M, et al. IL-6 blockade inhibits the induction of myelin antigen-specific Th17 cells and Th1 cells in experimental autoimmune encephalomyelitis. Proc Natl Acad Sci U S A 2008; 105: 9041–9046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Langley RG, Elewski BE, Lebwohl M, et al. Secukinumab in plaque psoriasis – results of two phase 3 trials. N Engl J Med 2014; 371: 326–338. [DOI] [PubMed] [Google Scholar]
- 86.Genovese MC, Durez P, Richards HB, et al. Efficacy and safety of secukinumab in patients with rheumatoid arthritis: a phase II, dose-finding, double-blind, randomised, placebo controlled study. Ann Rheum Dis 2013; 72: 863–869. [DOI] [PubMed] [Google Scholar]
- 87.McInnes IB, Sieper J, Braun J, et al. Efficacy and safety of secukinumab, a fully human anti-interleukin-17A monoclonal antibody, in patients with moderate-to-severe psoriatic arthritis: a 24-week, randomised, double-blind, placebo-controlled, phase II proof-of-concept trial. Ann Rheum Dis 2014; 73: 349–356. [DOI] [PubMed] [Google Scholar]
- 88.Lock C, Hermans G, Pedotti R, et al. Gene-microarray analysis of multiple sclerosis lesions yields new targets validated in autoimmune encephalomyelitis. Nat Med 2002; 8: 500–508. [DOI] [PubMed] [Google Scholar]
- 89.Ivanov II, McKenzie BS, Zhou L, et al. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 2006; 126: 1121–1133. [DOI] [PubMed] [Google Scholar]
- 90.Venturini M, Zanca A, Venturuzzo A, et al. Secukinumab for patients with plaque psoriasis affected by multiple sclerosis: a mini-review with a representative case report. J Eur Acad Dermatol Venereol 2020; 34: e110–e112. [DOI] [PubMed] [Google Scholar]
- 91.Di Tullio F, Odorici G, Lasagni C, et al. Combination treatment with secukinumab and dimethyl fumarate in a patient with psoriasis and recent diagnosis of multiple sclerosis. Dermatol Ther 2020; 33: e13943. [DOI] [PubMed] [Google Scholar]
- 92.Cortese A, Lucchetti R, Altobelli A, et al. Secukinumab may be a valid treatment option in patients with CNS demyelination and concurrent ankylosing spondylitis: report of two clinical cases. Mult Scler Relat Disord 2019; 35: 193–195. [DOI] [PubMed] [Google Scholar]
- 93.Assefa GT, Kaneko S, Oguro H, et al. Treatment of psoriasis and psoriatic arthritis with secukinumab after unsatisfactory response to ustekinumab in multiple sclerosis patient. J Dermatol 2019; 46: e112–e113. [DOI] [PubMed] [Google Scholar]
- 94.Diebold M, Muller S, Derfuss T, et al. A case of concomitant psoriasis and multiple sclerosis: secukinumab and rituximab exert dichotomous effects in two autoimmune conditions. Mult Scler Relat Disord 2019; 31: 38–40. [DOI] [PubMed] [Google Scholar]
- 95.Papp KA, Langley RG, Lebwohl M, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 52-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 2). Lancet 2008; 371: 1675–1684. [DOI] [PubMed] [Google Scholar]
- 96.Griffiths CE, Strober BE, van de Kerkhof P, et al. Comparison of ustekinumab and etanercept for moderate-to-severe psoriasis. N Engl J Med 2010; 362: 118–128. [DOI] [PubMed] [Google Scholar]
- 97.Sands BE, Sandborn WJ, Panaccione R, et al. Ustekinumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med 2019; 381: 1201–1214. [DOI] [PubMed] [Google Scholar]
- 98.Leonard JP, Waldburger KE, Goldman SJ. Prevention of experimental autoimmune encephalomyelitis by antibodies against interleukin 12. J Exp Med 1995; 181: 381–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Awasthi A, Riol-Blanco L, Jager A, et al. Cutting edge: IL-23 receptor gfp reporter mice reveal distinct populations of IL-17-producing cells. J Immunol 2009; 182: 5904–5908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.McGeachy MJ, Chen Y, Tato CM, et al. The interleukin 23 receptor is essential for the terminal differentiation of interleukin 17-producing effector T helper cells in vivo. Nat Immunol 2009; 10: 314–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Segal BM, Constantinescu CS, Raychaudhuri A, et al. Repeated subcutaneous injections of IL12/23 p40 neutralising antibody, ustekinumab, in patients with relapsing–remitting multiple sclerosis: a phase II, double-blind, placebo-controlled, randomised, dose-ranging study. Lancet Neurol 2008; 7: 796–804. [DOI] [PubMed] [Google Scholar]
- 102.Cravens PD, Hussain RZ, Miller-Little WA, et al. IL-12/IL-23p40 is highly expressed in secondary lymphoid organs and the CNS during all stages of EAE, but its deletion does not affect disease perpetuation. PLoS One 2016; 11: e0165248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Chang S, Chambers CJ, Liu FT, et al. Successful treatment of psoriasis with ustekinumab in patients with multiple sclerosis. Dermatol Online J 2015; 21: 13030/qt3bs971cr. [PubMed] [Google Scholar]
- 104.Schweckendiek W. [Treatment of psoriasis vulgaris]. Med Monatsschr 1959; 13: 103–104. [PubMed] [Google Scholar]
- 105.Schimrigk S, Brune N, Hellwig K, et al. Oral fumaric acid esters for the treatment of active multiple sclerosis: an open-label, baseline-controlled pilot study. Eur J Neurol 2006; 13: 604–610. [DOI] [PubMed] [Google Scholar]
- 106.Yadav SK, Soin D, Ito K, et al. Insight into the mechanism of action of dimethyl fumarate in multiple sclerosis. J Mol Med (Berl) 2019; 97: 463–472. [DOI] [PubMed] [Google Scholar]
- 107.Parodi B, Rossi S, Morando S, et al. Fumarates modulate microglia activation through a novel HCAR2 signaling pathway and rescue synaptic dysregulation in inflamed CNS. Acta Neuropathol 2015; 130: 279–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Baird L, Dinkova-Kostova AT. The cytoprotective role of the Keap1-Nrf2 pathway. Arch Toxicol 2011; 85: 241–272. [DOI] [PubMed] [Google Scholar]
- 109.Ghoreschi K, Bruck J, Kellerer C, et al. Fumarates improve psoriasis and multiple sclerosis by inducing type II dendritic cells. J Exp Med 2011; 208: 2291–2303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Altmeyer PJ, Matthes U, Pawlak F, et al. Antipsoriatic effect of fumaric acid derivatives. Results of a multicenter double-blind study in 100 patients. J Am Acad Dermatol 1994; 30: 977–981. [DOI] [PubMed] [Google Scholar]
- 111.Gold R, Kappos L, Arnold DL, et al. Placebo-controlled phase 3 study of oral BG-12 for relapsing multiple sclerosis. N Engl J Med 2012; 367: 1098–1107. [DOI] [PubMed] [Google Scholar]
- 112.Kappos L, Gold R, Miller DH, et al. Efficacy and safety of oral fumarate in patients with relapsing–remitting multiple sclerosis: a multicentre, randomised, double-blind, placebo-controlled phase IIb study. Lancet 2008; 372: 1463–1472. [DOI] [PubMed] [Google Scholar]
- 113.Casili G, Cordaro M, Impellizzeri D, et al. Dimethyl fumarate reduces inflammatory responses in experimental colitis. J Crohns Colitis 2016; 10: 472–483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Kourakis S, Timpani CA, de Haan JB, et al. Dimethyl fumarate and its esters: a drug with broad clinical utility? Pharmaceuticals (Basel) 2020; 13: 306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Tsivgoulis G, Papadimitropoulos GN, Katsanos AH, et al. Teaching NeuroImages: interferon-induced psoriasis flare in a multiple sclerosis case remits with dimethyl fumarate. Neurology 2017; 89: e188–e189. [DOI] [PubMed] [Google Scholar]
- 116.Zecca C, Caporro M, Adami M, et al. Fumaric acid esters in psoriasis and multiple sclerosis. Clin Exp Dermatol 2014; 39: 488–491. [DOI] [PubMed] [Google Scholar]
- 117.Corr M, Boyle DL, Ronacher LM, et al. Interleukin 1 receptor antagonist mediates the beneficial effects of systemic interferon beta in mice: implications for rheumatoid arthritis. Ann Rheum Dis 2011; 70: 858–863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.van Holten J, Reedquist K, Sattonet-Roche P, et al. Treatment with recombinant interferon-beta reduces inflammation and slows cartilage destruction in the collagen-induced arthritis model of rheumatoid arthritis. Arthritis Res Ther 2004; 6: R239–R249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.van Holten J, Pavelka K, Vencovsky J, et al. A multicentre, randomised, double blind, placebo controlled phase II study of subcutaneous interferon beta-1a in the treatment of patients with active rheumatoid arthritis. Ann Rheum Dis 2005; 64: 64–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Jabaily JA, Thompson JS. Effects of interferon beta-1B in rheumatoid arthritis: a case report. Arthritis Rheum 1997; 40: 1370. [DOI] [PubMed] [Google Scholar]
- 121.Alsalameh S, Manger B, Kern P, et al. New onset of rheumatoid arthritis during interferon beta-1B treatment in a patient with multiple sclerosis: comment on the case report by Jabaily and Thompson. Arthritis Rheum 1998; 41: 754. [DOI] [PubMed] [Google Scholar]
- 122.Altintas A, Alici Y, Melikoglu M, et al. Arthritis during interferon beta-1b treatment in multiple sclerosis. Mult Scler 2002; 8: 534–536. [DOI] [PubMed] [Google Scholar]
- 123.Nikolaus S, Rutgeerts P, Fedorak R, et al. Interferon beta-1a in ulcerative colitis: a placebo controlled, randomised, dose escalating study. Gut 2003; 52: 1286–1290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Musch E, Andus T, Kruis W, et al. Interferon-beta-1a for the treatment of steroid-refractory ulcerative colitis: a randomized, double-blind, placebo-controlled trial. Clin Gastroenterol Hepatol 2005; 3: 581–586. [DOI] [PubMed] [Google Scholar]
- 125.Pena-Rossi C, Schreiber S, Golubovic G, et al. Clinical trial: a multicentre, randomized, double-blind, placebo-controlled, dose-finding, phase II study of subcutaneous interferon-beta-la in moderately active ulcerative colitis. Aliment Pharmacol Ther 2008; 28: 758–767. [DOI] [PubMed] [Google Scholar]
- 126.Sprenger R, Sagmeister M, Offner F. Acute ulcerative colitis during successful interferon/ribavirin treatment for chronic hepatitis. Gut 2005; 54: 438–439; author reply 439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Rodrigues S, Magro F, Soares J, et al. Case series: ulcerative colitis, multiple sclerosis, and interferon-beta 1a. Inflamm Bowel Dis 2010; 16: 2001–2003. [DOI] [PubMed] [Google Scholar]
- 128.Schott E, Paul F, Wuerfel JT, et al. Development of ulcerative colitis in a patient with multiple sclerosis following treatment with interferon beta 1a. World J Gastroenterol 2007; 13: 3638–3640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Warabi Y, Matsumoto Y, Hayashi H. Interferon beta-1b exacerbates multiple sclerosis with severe optic nerve and spinal cord demyelination. J Neurol Sci 2007; 252: 57–61. [DOI] [PubMed] [Google Scholar]
- 130.Shimizu J, Hatanaka Y, Hasegawa M, et al. IFNβ-1b may severely exacerbate Japanese optic-spinal MS in neuromyelitis optica spectrum. Neurology 2010; 75: 1423–1427. [DOI] [PubMed] [Google Scholar]
- 131.Toussirot E, Bereau M, Bossert M, et al. Occurrence of psoriatic arthritis during interferon beta 1a treatment for multiple sclerosis. Case Rep Rheumatol 2014; 2014: 949317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Nestle FO, Nickoloff BJ. From classical mouse models of psoriasis to a spontaneous xenograft model featuring use of AGR mice. Ernst Schering Res Found Workshop2005; 50: 203–212. [DOI] [PubMed] [Google Scholar]
- 133.Gregorio J, Meller S, Conrad C, et al. Plasmacytoid dendritic cells sense skin injury and promote wound healing through type I interferons. J Exp Med 2010; 207: 2921–2930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Gui J, Gober M, Yang X, et al. Therapeutic elimination of the type 1 interferon receptor for treating psoriatic skin inflammation. J Invest Dermatol 2016; 136: 1990–2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.O’Sullivan S, Dev KK. Sphingosine-1-phosphate receptor therapies: advances in clinical trials for CNS-related diseases. Neuropharmacology 2017; 113: 597–607. [DOI] [PubMed] [Google Scholar]
- 136.Kappos L, Radue EW, O’Connor P, et al. A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. N Engl J Med 2010; 362: 387–401. [DOI] [PubMed] [Google Scholar]
- 137.Schaper K, Dickhaut J, Japtok L, et al. Sphingosine-1-phosphate exhibits anti-proliferative and anti-inflammatory effects in mouse models of psoriasis. J Dermatol Sci 2013; 71: 29–36. [DOI] [PubMed] [Google Scholar]
- 138.Jin J, Xue N, Liu Y, et al. A novel S1P1 modulator IMMH002 ameliorates psoriasis in multiple animal models. Acta Pharm Sin B 2020; 10: 276–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Mizushima T, Ito T, Kishi D, et al. Therapeutic effects of a new lymphocyte homing reagent FTY720 in interleukin-10 gene-deficient mice with colitis. Inflamm Bowel Dis 2004; 10: 182–192. [DOI] [PubMed] [Google Scholar]
- 140.Deguchi Y, Andoh A, Yagi Y, et al. The S1P receptor modulator FTY720 prevents the development of experimental colitis in mice. Oncol Rep 2006; 16: 699–703. [PubMed] [Google Scholar]
- 141.Yoshida Y, Mikami N, Matsushima Y, et al. Combination treatment with fingolimod and a pathogenic antigen prevents relapse of glucose-6-phosphate isomerase peptide-induced arthritis. Immun Inflamm Dis 2016; 4: 263–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Yoshida Y, Mikami N, Matsushima Y, et al. Functional mechanism(s) of the inhibition of disease progression by combination treatment with fingolimod plus pathogenic antigen in a glucose-6-phosphate isomerase peptide-induced arthritis mouse model. Biol Pharm Bull 2015; 38: 1120–1125. [DOI] [PubMed] [Google Scholar]
- 143.Krause A, D’Ambrosio D, Dingemanse J. Modeling clinical efficacy of the S1P receptor modulator ponesimod in psoriasis. J Dermatol Sci 2018; 89: 136–145. [DOI] [PubMed] [Google Scholar]
- 144.Vaclavkova A, Chimenti S, Arenberger P, et al. Oral ponesimod in patients with chronic plaque psoriasis: a randomised, double-blind, placebo-controlled phase 2 trial. Lancet 2014; 384: 2036–2045. [DOI] [PubMed] [Google Scholar]
- 145.Sandborn WJ, Feagan BG. Ozanimod treatment for ulcerative colitis. N Engl J Med 2016; 375: e17. [DOI] [PubMed] [Google Scholar]
- 146.Cherwinski HM, Byars N, Ballaron SJ, et al. Leflunomide interferes with pyrimidine nucleotide biosynthesis. Inflamm Res 1995; 44: 317–322. [DOI] [PubMed] [Google Scholar]
- 147.Kaltwasser JP, Nash P, Gladman D, et al. Efficacy and safety of leflunomide in the treatment of psoriatic arthritis and psoriasis: a multinational, double-blind, randomized, placebo-controlled clinical trial. Arthritis Rheum 2004; 50: 1939–1950. [DOI] [PubMed] [Google Scholar]
- 148.Osiri M, Deesomchok U, Tugwell P. Disease activity and functional changes of RA patients receiving different DMARDs in clinical practice. Clin Rheumatol 2006; 25: 721–727. [DOI] [PubMed] [Google Scholar]
- 149.Son M, McEwan L, Ubaidat M, et al. Lymphocytic colitis in the setting of teriflunomide use for relapsing multiple sclerosis. Mult Scler 2020; 26: 1801–1803. [DOI] [PubMed] [Google Scholar]
- 150.Esfahani NZ, von Geldern G, Romba MC, et al. Inflammatory colitis associated with Teriflunomide. Mult Scler Relat Disord 2020; 46: 102480. [DOI] [PubMed] [Google Scholar]
- 151.Dereure O, Camu W. Teriflunomide-induced psoriasiform changes of fingernails: a new example of paradoxical side effect? Int J Dermatol 2017; 56: 1479–1481. [DOI] [PubMed] [Google Scholar]
- 152.Negrotto L, Correale J. Palmar pustular psoriasis associated with teriflunomide treatment. Mult Scler Relat Disord 2019; 27: 400–402. [DOI] [PubMed] [Google Scholar]
- 153.Baldin E, Lugaresi A. Ponesimod for the treatment of relapsing multiple sclerosis. Expert Opin Pharmacother 2020; 21: 1955–1964. [DOI] [PubMed] [Google Scholar]
- 154.Khoury SJ, Rochon J, Ding L, et al. ACCLAIM: a randomized trial of abatacept (CTLA4-Ig) for relapsing–remitting multiple sclerosis. Mult Scler 2017; 23: 686–695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Herfarth HH. Methotrexate for inflammatory bowel diseases – new developments. Dig Dis 2016; 34: 140–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Freeman HJ. Colitis associated with biological agents. World J Gastroenterol 2012; 18: 1871–1874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Mizoguchi E, Mizoguchi A, Preffer FI, et al. Regulatory role of mature B cells in a murine model of inflammatory bowel disease. Int Immunol 2000; 12: 597–605. [DOI] [PubMed] [Google Scholar]
- 158.Aharoni R, Kayhan B, Brenner O, et al. Immunomodulatory therapeutic effect of glatiramer acetate on several murine models of inflammatory bowel disease. J Pharmacol Exp Ther 2006; 318: 68–78. [DOI] [PubMed] [Google Scholar]
- 159.Yao Y, Han W, Liang J, et al. Glatiramer acetate ameliorates inflammatory bowel disease in mice through the induction of Qa-1-restricted CD8+ regulatory cells. Eur J Immunol 2013; 43: 125–136. [DOI] [PubMed] [Google Scholar]
- 160.Aharoni R, Sonego H, Brenner O, et al. The therapeutic effect of glatiramer acetate in a murine model of inflammatory bowel disease is mediated by anti-inflammatory T-cells. Immunol Lett 2007; 112: 110–119. [DOI] [PubMed] [Google Scholar]
- 161.Papp KA, Menter A, Strober B, et al. Efficacy and safety of tofacitinib, an oral Janus kinase inhibitor, in the treatment of psoriasis: a phase 2b randomized placebo-controlled dose-ranging study. Br J Dermatol 2012; 167: 668–677. [DOI] [PubMed] [Google Scholar]
- 162.Lee EB, Fleischmann R, Hall S, et al. Tofacitinib versus methotrexate in rheumatoid arthritis. N Engl J Med 2014; 370: 2377–2386. [DOI] [PubMed] [Google Scholar]
- 163.Sandborn WJ, Su C, Sands BE, et al. Tofacitinib as induction and maintenance therapy for ulcerative colitis. N Engl J Med 2017; 376: 1723–1736. [DOI] [PubMed] [Google Scholar]
- 164.van de Kerkhof PC, Reich K, Kavanaugh A, et al. Physician perspectives in the management of psoriasis and psoriatic arthritis: results from the population-based multinational assessment of psoriasis and psoriatic arthritis survey. J Eur Acad Dermatol Venereol 2015; 29: 2002–2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Zhou Y, Leng X, Luo S, et al. Tolerogenic dendritic cells generated with tofacitinib ameliorate experimental autoimmune encephalomyelitis through modulation of Th17/Treg balance. J Immunol Res 2016; 2016: 5021537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Massoud F, Ismail II, Al-Hashel JY, et al. CNS demyelination during tofacitinib therapy: first report. Mult Scler Relat Disord 2020; 46: 102568. [DOI] [PubMed] [Google Scholar]
- 167.Benveniste EN, Liu Y, McFarland BC, et al. Involvement of the Janus kinase/signal transducer and activator of transcription signaling pathway in multiple sclerosis and the animal model of experimental autoimmune encephalomyelitis. J Interferon Cytokine Res 2014; 34: 577–588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Magliozzi R, Marastoni D, Calabrese M. The BAFF/APRIL system as therapeutic target in multiple sclerosis. Expert Opin Ther Targets 2020; 24: 1135–1145. [DOI] [PubMed] [Google Scholar]
- 169.Vincent FB, Saulep-Easton D, Figgett WA, et al. The BAFF/APRIL system: emerging functions beyond B cell biology and autoimmunity. Cytokine Growth Factor Rev 2013; 24: 203–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Kappos L, Hartung HP, Freedman MS, et al. Atacicept in Multiple Sclerosis (ATAMS): a randomised, placebo-controlled, double-blind, phase 2 trial. Lancet Neurol 2014; 13: 353–363. [DOI] [PubMed] [Google Scholar]
- 171.Baker D, Pryce G, James LK, et al. Failed B cell survival factor trials support the importance of memory B cells in multiple sclerosis. Eur J Neurol 2020; 27: 221–228. [DOI] [PubMed] [Google Scholar]
- 172.Richez C, Truchetet ME, Schaeverbeke T, et al. Atacicept as an investigated therapy for rheumatoid arthritis. Expert Opin Investig Drugs 2014; 23: 1285–1294. [DOI] [PubMed] [Google Scholar]
- 173.Liu YT, Ding HH, Lin ZM, et al. A novel tricyclic BTK inhibitor suppresses B cell responses and osteoclastic bone erosion in rheumatoid arthritis. Acta Pharmacol Sin. Epub ahead of print 13January2021. DOI: 10.1038/s41401-020-00578-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Torke S, Weber MS. Inhibition of Bruton s tyrosine kinase as a novel therapeutic approach in multiple sclerosis. Expert Opin Investig Drugs 2020; 29: 1143–1150. [DOI] [PubMed] [Google Scholar]



