Loading...
 
Mediterr J Rheumatol 2022;33(Suppl 1):126-36
Inflammatory Bowel Disease-related Spondyloarthritis: The Last Unexplored Territory of Rheumatology
Authors Information

First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece

Abstract

The Spondyloarthritides (SpA) are a group of chronic inflammatory diseases affecting the spine, peripheral joints and entheses, as well as extra-skeletal structures, including the gastrointestinal tract. On the other hand, inflammatory bowel disease (IBD), either Crohn’s disease or ulcerative colitis, often affects extra-intestinal sites, including the axial and/or peripheral skeleton. IBD-related arthritis is the type of SpA that occurs in patients affected by IBD, with an incidence up to 50% during the IBD course. Although both manifestations are apparently the result of a common pathogenetic process, physicians often fail to recognize the disease in its entirety: thus, IBD-SpA is managed as two separate diseases, a musculoskeletal and a gastrointestinal one, with a profound impact on patient quality of life. Moreover, the specialty of the treating physician determines the clinical and laboratory tools for disease assessment, which, in turn, guide treatment decisions that may overlook either affected system or even act in the opposite direction. Raising awareness of the intestinal and musculoskeletal manifestations among rheumatologists and gastroenterologists will lead to earlier diagnosis and a multidisciplinary approach, particularly regarding pharmacologic treatments. Given the lack of trial evidence on immunomodulatory drugs in IBD-SpA it is imperative for researchers in both medical disciplines to join efforts, in order to determine referral strategies, appropriate composite measures for disease assessment, treatment algorithms and therapeutic targets.


Cite this article as: Zioga N, Kogias D, Lampropoulou V, Kafalis N, Papagoras C. Inflammatory Bowel Disease-related Spondyloarthritis: The Last Unexplored Territory of Rheumatology. Mediterr J Rheumatol 2022;33(Suppl 1):126-36.

Article Submitted: 29 May 2021; Article Accepted: 20 Jun 2021; Available Online: 15 Apr 2022

https://doi.org/10.31138/mjr.33.1.126

This work is licensed under a Creative Commons Attribution 4.0 International License (CC-BY).

©Zioga N, Kogias D, Lampropoulou V, Kafalis N, Papagoras C.

Full Text

INTRODUCTION

It has been over 90 years since the first report linking ulcerative colitis with arthritis,1 and over 60 years, it became evident that the musculoskeletal involvement accompanying ulcerative colitis (UC) and Crohn’s disease (CD) has much in common with ankylosing spondylitis (AS).2,3 Following the introduction of the unifying concept of spondyloarthritis (SpA),4 the inflammatory bowel disease (IBD) has been regarded as a hallmark extraskeletal feature of SpA and has been incorporated in all sets of SpA classification criteria, including the most recent one proposed by the Assessment of Spondyloarthritis International Society (ASAS).5-7

The recognition of the link between intestinal inflammation and joint disease has led to the introduction of the term “enteropathic arthritis”. Although medical textbooks generally reserve this term for the musculoskeletal disease accompanying CD and UC, enteropathic arthritis could mean any type of arthritis associated with intestinal disease. Indeed, the term has also been used to denote joint diseases within the SpA spectrum, such as reactive arthritis, or outside of it, such as arthritis in Whipple’s disease, celiac disease or following intestinal bypass surgery.8 It is remarkable that in the 10th revision of the International Classification of Diseases (ICD) that is still broadly used, there are 3 codes for enteropathic arthropathy, ie, “in Crohn disease” (M07.4), “in ulcerative colitis” (M07.5) and “other enteropathic arthropathies” (M07.6). To make things more complicated, in the latest 11th ICD revision, the term “enteropathic arthropathies” is completely eliminated and the CD- and UC-related arthritides are coded with the term “Other specified reactive arthropathies” (FA11.Y). Although this appears a minor detail, it actually preserves the century-old belief that IBD-related arthritis is a mere musculoskeletal reflection of the intestinal inflammation and, therefore, the treatment of IBD is sufficient to address joint disease, as well. This and other misconceptions are probably the reason why IBD-related SpA, perhaps the most appropriate term, has been largely ignored by rheumatologic clinical and translational research, in contrast to its skin inflammatory disease counterpart, ie, psoriatic arthritis (PsA).

 

PREVALENCE AND TYPES OF MUSCULOSKELETAL INVOLVEMENT

The introduction of the Amor5 and European Spondylarthropathy Study Group (ESSG) criteria,in the early 90’s, for the first time, allowed researchers to formally classify IBD patients with musculoskeletal symptoms as suffering from SpA. In 2009, the ASAS criteria introduced some innovations in the SpA concept that apply to IBD-related SpA as well: the clinical manifestations are distinguished in axial and peripheral, while axial SpA is further divided in radiographic and non-radiographic depending on whether sacroiliac damage is evident on plain radiographs.7,9 However, most studies on the epidemiology of IBD-related SpA used the older criteria.10 Hence, the overall prevalence of SpA manifestations in IBD patients was estimated between 17% -62% and the prevalence of SpA according to the ESSG criteria between 5% -45.7%.11 More recently, in a Norwegian cohort of IBD patients (IBSEN cohort), 36% fulfilled the ASAS criteria for SpA, of whom 8% for axial SpA and 28% for peripheral SpA.12

 

Axial disease

Axial involvement in IBD has been reported under various definitions, such as AS, sacroiliitis and inflammatory back pain.  According to a meta-analysis, the pooled prevalence of AS in IBD is 3% (95% confidence interval [CI] 2-4%).10 Conversely, in a meta-analysis of extra-articular manifestations of AS, the prevalence of IBD was estimated at 6.8% (95% CI 6.1% - 7.7%).13 Sacroiliitis in IBD patients is more frequent, with a pooled prevalence of 10% (95% CI 8-12%), which may be either clinical (pooled prevalence 8%), or subclinical (pooled prevalence 11%).10 This variation might be due to IBD treatment with tumour necrosis factor-α (TNFα) inhibitors, which improve the spinal symptoms, but may not equally affect radiographic damage. Finally, the most frequent axial manifestation of IBD is inflammatory back pain (IBP), which has been reported in 5.2-42% of patients.11 As in the rest of axial SpA, a possible explanation might be that IBD-related SpA begins with inflammatory back pain followed over the years, at least in a proportion of patients, by structural damage allowing it to be diagnosed as radiographic axial SpA (AS). Although in this regard more sensitive imaging modalities would reveal a higher prevalence of sacroiliitis in the IBD population, sacroiliitis has been identified at similar proportions with conventional radiography (12%), computed tomography (CT) (15%) and magnetic resonance imaging (MRI) (10%) (10). Concerning the type of IBD, it appears that axial manifestations occur slightly more often in CD than UC.10

 

Peripheral disease

Peripheral arthritis, enthesitis and dactylitis are another type of musculoskeletal involvement in IBD-related SpA. According to the above-mentioned meta-analysis the pooled prevalence of peripheral arthritis in IBD is 13% (95% CI 12–15%) with a slight predominance in CD (15%) than UC (12%).10 The incidence of peripheral arthritis was also assessed in the IBSEN cohort of IBD patients who were followed over 20 years. The authors reported that 76 out of 441 patients (17.2%) had ever manifested pure peripheral arthritis that could be attributed to IBD. There was a significant predominance of females (67%), while there was no difference in the prevalence of CD-related and UC-related peripheral arthritis. Finally, another 19 patients (4.3%) had at least one instance of enthesitis or dactylitis.14 It should be noted, though, that there is a high variability in the rates of enthesitis across studies, ranging from 1-54% with no clear difference between CD and UC.10,11 In contrast, dactylitis has been reported in less than 5% of patients with either CD or UC.10

Concerning patients’ perceptions of the impact of the disease on their quality of life, in the IBSEN cohort the presence of arthralgia or back pain was independently associated with worse scores across all domains (except for mental health) of the Short Form 36 (SF-36) questionnaire, as well as with higher levels of fatigue and chronic fatigue.12 Moreover, in an Italian cohort of IBD-related SpA, the variables that had a significant positive correlation with function, as assessed with the Health Assessment Questionnaire (HAQ), were the Bath Ankylosing Spondylitis Functional Index (BASFI) and the AS Disease Activity Index-C-Reactive Protein (ASDAS-CRP), but not measures of intestinal disease activity, such as the partial Mayo score, the Crohn’s Disease Activity Index (CDAI) or the IBD Questionnaire (IBDQ).15 It is, thus, obvious, that musculoskeletal manifestations are frequent in patients with IBD causing considerable pain and impairment in their quality of life, independently to that inflicted by the IBD itself.

PATHOPHYSIOLOGICAL LINKS BETWEEN GUT AND SKELETAL INFLAMMATION

The current concept of the pathogenesis of SpA holds that the initial immunological challenge occurs at the level of intestinal mucosa (or the skin in the case of PsA). It is assumed that a particular genetic background allows for an aberrant gut immune response against rather common insults, which eventually spreads and persists at musculoskeletal sites.16

Indeed, several studies in which axial SpA patients were screened for gut inflammation by ileocolonoscopy and biopsy or endoscopic capsule raised the prevalence of subclinical gut inflammation to more than 50% of patients with axial SpA, almost 10-fold higher than the reported co-existence of clinical IBD and AS.17-19 Regarding peripheral arthritis, Orchard et al. divided IBD-related peripheral arthritis into 2 types: type 1, an oligoarthritis involving large joints of the legs, which typically occurs during periods of active gut disease and wanes off within weeks; and type 2, a chronic arthritis which may involve several joints, including small ones of the hands, and lacks a clear association with the activity of the intestinal disease. In 4-14% of patients with either type, the arthritis occurred before IBD diagnosis, but it is uncertain whether it predated the actual onset of IBD.20 These observations suggest that in some IBD patients the arthritis may be a remote repercussion of active gut inflammation, much like reactive arthritis. Importantly, though, in a proportion of IBD patients, arthritis seems to be a separate manifestation of the disease, which, once triggered, follows an independent course from the bowel disease.

From a pathophysiological aspect, several genetic loci related to immune function, cytokine signalling and major histocompatibility complex (MHC) proteins have been associated with either SpA and IBD, with plenty of them being common for both diseases.21 Moreover, environmental stresses such a smoking and, particularly, intestinal dysbiosis have been implicated in the pathogenesis of both IBD and SpA, while mechanical forces are related solely to SpA.16,22,23

A possible mechanism linking genes, microbiota, intestinal and joint disease is exemplified by the human leukocyte antigen B27 (HLA B27)/β2-microglobulin transgenic rat model of human SpA. In this model, immunological changes in the gut, such as production of interleukin 1β (IL-1β) and TNFα occur early, followed by an increased expression of interferon γ (IFNγ) and type 3 immunity cytokines (IL-23, IL-17). Subsequently, dysbiotic changes and intestinal inflammation develop prior to the ultimate onset of clinical arthritis.24,25 In healthy humans, carriage of HLA B27 has also been associated with intestinal microbiota alterations,26 while intestinal dysbiosis and subclinical gut inflammation is a feature of AS.27 Innate mechanisms, such as autophagy, are involved at the intestinal level both in the subclinical gut inflammation of AS, as well as in active UC.28,29 The activation of type-3 immunity in the gut involves local upregulation of IL-23 and expansion of IL-17-producing type 3 innate lymphoid cells (ILC-3) cells, which circulate and potentially relocate to skeletal sites.30,31 Moreover, in a rat model of SpA, IL-23 may activate cells of the innate immunity at the entheses to produce IL-17 and IL-22, both important downstream effector cytokines in the pathogenesis of SpA.32 Recent research has demonstrated that normal human entheses also harbour cells of the innate immunity (ILC-3 and Tγ/δ lymphocytes) that are able to produce IL-17 both in an IL-23-dependent and independent manner.33,34

It is possible that IBD and SpA share a common initial sequence of events that unfold at the gut mucosa. However, differences in the genetic background, environmental and other as-yet unidentified factors may determine whether the inflammatory process expands and persists locally evolving to clinical IBD and/or relocates and establishes itself at entheseal sites giving rise to clinical SpA.

 

DIAGNOSIS

The key to early diagnosis of IBD-SpA is a high level of suspicion of rheumatologists for gastrointestinal symptoms in SpA patients and of gastroenterologists for musculoskeletal symptoms in IBD patients. Several questionnaires35-37 and screening tools38 have been proposed to help gastroenterologists identify musculoskeletal involvement in IBD patients and vice-versa, although their performance in clinical practice remains to be validated. Moreover, expert panels including rheumatologists and gastroenterologists have pointed some signs or symptoms that should be regarded as “red flags” suggestive of IBD or SpA and should prompt for further investigation.11,39-43 (Table 1) On the other hand, simple laboratory studies may be ambiguous and cause diagnostic delays. Indeed, among first-line laboratory tests, only pelvis X-rays carry a high positive predictive value for the diagnosis of sacroiliitis.44 In this regard, gastroenterologists requesting an abdominal CT should also look at the sacroiliac joints for structural damage due to sacroiliitis. Other studies, such as C-reactive protein (CRP) and HLA B27 are poorly sensitive or specific.45-48 Faecal or serum calprotectin has also been associated with subclinical gut inflammation in SpA patients,49-52 although faecal calprotectin levels may be influenced by non-steroidal anti-inflammatory drug use, which is common in SpA patients.50 However, in most cases the confirmation of IBD or SpA requires advanced or interventional procedures, such as musculoskeletal MRI, arthrocentesis and synovial fluid examination, gastrointestinal imaging, endoscopy or biopsy. The indication to perform and the interpretation of those tests are determined by the specialist, which highlights the importance of symptom-based referral and the gastroenterologist-rheumatologist collaboration.

Although the ASAS criteria may be helpful in identifying patients with SpA,7 their validity has not been assessed in IBD-SpA. In fact, there are some peculiarities in this disease that potentially affect the weight of some items and the sensitivity and specificity of the whole system. These include the lower prevalence of HLA B27 in IBD-SpA compared to classic AS,46-48 the relative contra-indication of non-steroidal anti-inflammatory drugs (NSAIDs) in patients with IBD53 and the lack of specificity of elevated serum CRP, as it may be due to active intestinal inflammation, as well.


Table 1. Proposed “red flags” to consider further investigation for concomitant IBD or SpA.
 

 

TREATMENT CONSIDERATIONS

Physicians treating patients with IBD-related SpA face two major challenges: although there are plenty of drugs approved separately for the treatment of axial SpA, PsA (a model for peripheral SpA) and IBD, there is almost complete lack of evidence concerning the efficacy of those treatments for the combined disease. Moreover, some of those drugs occasionally have contrasting effects on either disease aspect (Table 2).


Table 2. Overview of the effects of drugs approved for the treatment of IBD and SpA across the main disease manifestations.
 


NSAIDs are the first-line treatment for axial disease, enthesitis, dactylitis and often for peripheral arthritis according to recent recommendations.54,55 However, in a population study their use was associated with a dose-dependent risk for incident IBD.56 In IBD patients, NSAIDs have been associated with an increase in disease activity, particularly at high-doses and in patients with CD and colonic involvement.57,58 On the other hand, a couple of trials showed that selective cyclooxygenase-2 (COX-2) inhibitors (etoricoxib, celecoxib) in IBD patients was not associated with a greater risk for IBD exacerbation than placebo in the short term.59,60 Moreover, in a meta-analysis, the pooled proportion of IBD flare associated with selective COX-2 inhibitors was 14%, with most gastrointestinal symptoms occurring over the first few days to weeks of treatment.61 Summarizing, although NSAIDs, particularly selective COX-2 inhibitors, may be used for a short time as a symptomatic treatment in patients with quiescent IBD, their long-term safety is a concern and, thus, they are the least attractive treatment option for the chronic musculoskeletal manifestations of IBD-related SpA.62

Glucocorticoids (GC), either topically (oral budesonide) or systemically acting, are an effective therapy for inducing remission in CD,63,64 although they are not effective for maintaining remission.65 Systemic GC are also appropriate for patients with moderate to severe UC and for those with mild disease not responding to mesalazine. On the other hand, there is little place for GCs in the treatment of SpA. Although a trial of high dose oral GC in axial SpA showed a modest effect,66 long-term GC treatment of axial SpA is discouraged.54,55 Short-term low-dose oral GCs may be used in case of peripheral arthritis, while local GC injections may be used for the treatment of sacroiliitis, peripheral arthritis or enthesitis.54,55,67

Conventional immunomodulatory drugs used either for SpA or IBD include aminosalicylate compounds, methotrexate, leflunomide and azathioprine. Concerning aminosalicylates, both sulfasalazine and mesalazine are effective in UC, but their benefit in CD has not been well documented.68 Moreover, as mesalazine is better tolerated than sulfasalazine,69 most gastroenterologists would probably favour the former over the latter. This comes in contrast to the rheumatological practice, since only sulfasalazine may have some effect in peripheral arthritis of SpA, for which it is recommended.54,55,67,70,71 Conversely, published evidence does not support any benefit of sulfasalazine in reducing pain, disease activity, radiographic progression or improving physical function and spinal mobility in axial SpA.72,73

While methotrexate is an established treatment option for CD,74 a couple of randomised trials showed no significant benefit over placebo in UC.75,76 Concerning axial SpA the evidence supporting its use is very poor77,78 and, therefore, it is may be considered only for patients who cannot tolerate other drugs with proven efficacy.54,55 On the other hand, methotrexate is the first-line treatment for peripheral arthritis of PsA.67 Similarly, although leflunomide is efficacious in treating PsA,79 it is not efficacious in axial SpA80 while limited data for the treatment of IBD show some effectiveness in IBD-related joint pain.81,82 Finally, azathioprine, an immunosuppressant beneficial for both CD and UC83,84 has no place in the treatment of SpA manifestations.

Considering biologics, there are only few small open label studies assessing their efficacy for the treatment of both aspects of IBD-SpA.85-87 In fact, all randomized clinical trials of biologics in axial SpA excluded patients with active IBD.88-94 On the other hand, the published evidence on the treatment of peripheral SpA is dominated by trials in PsA, with only a few studies of adalimumab and golimumab in non-psoriatic peripheral SpA. In those studies, though, the proportion of patients with clinical IBD was too low to allow for meaningful conclusions.95-98 Conversely, no randomized trial of targeted treatments for CD and/or UC reported rheumatological outcomes.99-108

For over a decade TNFα blockade has been the only common second-line targeted treatment for both SpA and IBD. However, direct clinical evidence on the efficacy of TNF inhibitors across both domains of IBD-SpA is very limited. A trial of golimumab and infliximab showed efficacy on both aspects of the disease,85,86 while, etanercept is efficacious only for the musculoskeletal, but not gut involvement.87 Finally, the various currently available TNF inhibitors differ in terms of approved indications and dosages between SpA and IBD. Indeed, golimumab is not approved for CD, while certolizumab pegol is approved for CD in the US, but not in the EU. Moreover, dosages may differ between rheumatological and gastroenterological indications, with IBD generally requiring higher dosages (Tables 2 and 3).


Table 3. Dosages of targeted treatments approved for AxSpA/PsA, Crohn’s disease and Ulcerative Colitis.
 


Among biologics with alternative modes of action, the IL-12/23 blocker ustekinumab is effective and has been approved for the treatment of CD, UC and peripheral arthritis, enthesitis and dactylitis of PsA.105,108-110 Moreover, a retrospective study of patients with psoriasis or PsA and concomitant IBD treated with ustekinumab for the psoriatic disease showed a benefit for IBD as well.111 However, ustekinumab failed to show efficacy in the treatment of axial SpA,112 while there are no data on the treatment of non-psoriatic peripheral SpA.  Secukinumab is an IL-17 inhibitor which is effective for the treatment of axial SpA, as well as PsA.93,113,114 When tried in CD, though, a high rate of adverse events, mainly fungal infections, were reported.115 Moreover, there have been several reports of new onset or flaring IBD in patients with SpA, PsA, psoriasis or IBD treated with IL-17 blockers (secukinumab, ixekizumab, brodalumab). A class effect has been suspected and, therefore, IL-17 inhibitors are not currently recommended for patients with known IBD.116 However, long-term trial data, as well as meta-analyses showed a low risk of new onset IBD in patients with SpA, PsA or psoriasis treated with IL-17 blockers.117,118

The α4β7 integrin blocker vedolizumab is considered gut-specific and is used for the treatment of both CD and UC. A post-hoc analysis of the GEMINI trials of vedolizumab in IBD focusing on joint symptoms showed a lower risk for new-onset joint symptoms in CD, but not in UC.119 However, several case series have been published reporting new-onset or flaring musculoskeletal manifestations in IBD patients treated with vedolizumab.120,121 In a large cohort of IBD patients treated with vedolizumab, an improvement of joint symptoms was noted in 45% of patients who had such symptoms at baseline. The benefit was greatest in patients with recent-onset articular manifestations and those who attained IBD remission under vedolizumab. On the other hand, 14% of patients, mainly with CD and pre-existing AS, developed new onset joint symptoms under vedolizumab.122 These seemingly paradoxical results may actually reflect the potential different mechanisms of arthritis in IBD: a “reactive” mechanism in which suppressing bowel inflammation also benefits the extraintestinal manifestations; and an “autonomous” mechanism, in which once joint inflammation begins, it propagates independently of IBD activity.

Finally, tofacitinib has been approved for UC123,124 and PsA,125,126 while it has shown efficacy for the treatment of AS.127 However, there are almost no data concerning response of joint disease in IBD.128 Moreover, compared to PsA, treatment of UC requires higher doses of tofacitinib (Table 3) which have been associated with an increased risk of cardiovascular thromboembolic events.129

Last but not least, in recent years there is a significant interest in an unexplored field of treatment, the combination therapy. Multidrug therapy seems promising, but concerns for potential adverse events are high. There have been treatment considerations for combining traditional DMARDs with biological DMARDs or two biological DMARDs.130-134

Apart from the lack of single drug trials, there is very scarce evidence on how to start or escalate treatments in IBD-SpA. There is only a single investigator-initiated strategy trial involving both gastroenterologists and rheumatologists for the care of patients with IBD-SpA. In this study, treatment decisions were made based on the disease state over 3 domains (intestinal, axial or peripheral skeletal) according to which the patient would receive conventional synthetic disease-modifying anti-rheumatic drugs (DMARDs), or adalimumab. Notably, adalimumab was prescribed at the highest dosage, as approved for IBD. The study showed that this approach was associated with a significant improvement after 6 and 12 months in all outcomes measured, including BASDAI, ASDAS-CRP, CDAI, partial Mayo score, BASFI, HAQ, SF-36 and IBDQ.15 Although this was a small (N=52) and uncontrolled observational study, it also highlighted the value of collaborative work among physicians with different specialties for the treatment of IBD-SpA.

As evident from the above (Table 2) there is only a limited number of advanced treatments that appear safe and effective for all manifestations of IBD-SpA. In real world, though, it is not unusual for patients to have failed them all, making the treatment of IBD-SpA a real challenge for rheumatologists and gastroenterologists. There have been several cases of combining targeted drugs to treat IBD and SpA with often positive results.130-134 The main concern, though, is the long-term safety of combining immunomodulatory treatments, which should be addressed in clinical trials and long-term observational studies.

 

CONCLUSIONS

Inflammatory bowel disease-related spondyloarthritis is a common and debilitating disease that has not attracted equal scientific and medical attention. It is most important that rheumatologists and gastroenterologists become aware of both aspects of the disease, in order to actively seek for symptoms that will prompt further investigation and early diagnosis. Given the lack of scientific evidence, collaboration between physicians of both specialties is necessary, so that patients receive the appropriate treatments at the optimal dosages for their manifestations and are followed up properly. Regarding research, apart from studying the underlying pathophysiology, it is time that clinical trials include patients with both manifestations, so that the effect of existing and upcoming treatments is evaluated directly and not extrapolated from trials in Rheumatology and Gastroenterology alone. To this end, composite measures of gut and musculoskeletal disease activity should be developed and validated, in order to be used in trials, as well as in every day clinical practice. As this last rheumatological domain is gradually brought to light, it appears that there is still immense room for research and improvement in the care of those patients.

 

CONFLICT OF INTEREST

The authors declare no conflict of interest.

References
  1. Bargen JA. Complications and Sequelae of Chronic Ulcerative Colitis. Ann Intern Med 1929 Oct 1;3(4):335-52.
  2. McEwen C, Lingg C, Kirsner JB, Spencer JA. Arthritis accompanying ulcerative colitis. Am J Med 1962 Dec;33(6):923-41.
  3. Ansell BM, Wigley RAD. Arthritic Manifestations in Regional Enteritis. Ann Rheum Dis 1964 Jan 1;23(1):64-72.
  4. Wright V. Seronegative polyarthritis: a unified concept. Arthritis Rheum 1978;21(6):619-33.
  5. Amor B, Dougados M, Mijiyawa M. [Criteria of the classification of spondylarthropathies]. Rev Rhum Mal Osteoartic 1990 Feb;57(2):85-9.
  6. Dougados M, van der Linden S, Juhlin R, Huitfeldt B, Amor B, Calin  a, et al. The European Spondylarthropathy Study Group preliminary criteria for the classification of spondylarthropathy. Arthritis Rheum 1991 Oct;34(10):1218-27.
  7. Rudwaleit M, van der Heijde D, Landewé R, Akkoc N, Brandt J, Chou CT, et al. The Assessment of SpondyloArthritis International Society classification criteria for peripheral spondyloarthritis and for spondyloarthritis in general. Ann Rheum Dis 2011 Jan;70(1):25-31.
  8. Veys EM, Mielants H. Enteropathic arthritis, uveitis, Whipple’s disease, and miscellaneous spondyloarthropathies. Curr Opin Rheumatol 1993 Jul;5(4):420-7.
  9. Rudwaleit M, van der Heijde D, Landewé R, Listing J, Akkoc N, Brandt J, et al. The development of Assessment of SpondyloArthritis international Society classification criteria for axial spondyloarthritis (part II): validation and final selection. Ann Rheum Dis 2009 Jun;68(6):777-83.
  10. Karreman MC, Luime JJ, Hazes JMW, Weel AEAM. The Prevalence and Incidence of Axial and Peripheral Spondyloarthritis in Inflammatory Bowel Disease: A Systematic Review and Meta-analysis. J Crohns Colitis 2017 May 1;11(5):631-42.
  11. Salvarani C, Fries W. Clinical features and epidemiology of spondyloarthritides associated with inflammatory bowel disease. World J Gastroenterol 2009;15(20):2449-55.
  12. Ossum AM, Palm Ø, Cvancarova M, Bernklev T, Jahnsen J, Moum B, et al. The Impact of Spondyloarthritis and Joint Symptoms on Health-Related Quality of Life and Fatigue in IBD Patients. Results from a Population-Based Inception Cohort (20-Year Follow-up in the Ibsen Study). Inflamm Bowel Dis 2020;26(1):114-24.
  13. Stolwijk C, van Tubergen A, Castillo-Ortiz JD, Boonen A. Prevalence of extra-articular manifestations in patients with ankylosing spondylitis: a systematic review and meta-analysis. Ann Rheum Dis 2015 Jan 1;74(1):65-73.
  14. Ossum AM, Palm Ø, Cvancarova M, Solberg IC, Vatn M, Moum B, et al. Peripheral arthritis in patients with long-term inflammatory bowel disease. Results from 20 years of follow-up in the IBSEN study. Scand J Gastroenterol 2018 Nov 2;53(10-11):1250-6.
  15. Luchetti MM, Benfaremo D, Ciccia F, Bolognini L, Ciferri M, Farinelli A, et al. Adalimumab efficacy in enteropathic spondyloarthritis: A 12-mo observational multidisciplinary study. World J Gastroenterol 2017 Oct 21;23(39):7139-49.
  16. Paine A, Ritchlin CT. Targeting the interleukin-23/17 axis in axial spondyloarthritis. Curr Opin Rheumatol 2016 Jul;28(4):359-67.
  17. Smale S, Natt RS, Orchard TR, Russell AS, Bjarnason I. Inflammatory bowel disease and spondylarthropathy. Arthritis Rheum 2001 Dec;44(12):2728-36.
  18. Van Praet L, Van den Bosch FE, Jacques P, Carron P, Jans L, Colman R, et al. Microscopic gut inflammation in axial spondyloarthritis: a multiparametric predictive model. Ann Rheum Dis 2013 Mar 8;72(3):414-7.
  19. Kopylov U, Starr M, Watts C, Dionne S, Girardin M, Seidman EG. Detection of Crohn Disease in Patients with Spondyloarthropathy: The SpACE Capsule Study. J Rheumatol 2018 Apr;45(4):498-505.
  20. Orchard TR, Wordsworth BP, Jewell DP. Peripheral arthropathies in inflammatory bowel disease: their articular distribution and natural history. Gut 1998 Mar;42(3):387-91.
  21. Baeten D, Breban M, Lories R, Schett G, Sieper J. Are spondylarthritides related but distinct conditions or a single disease with a heterogeneous phenotype? Arthritis Rheum 2013 Jan 27;65(1):12-20.
  22. Nishida A, Inoue R, Inatomi O, Bamba S, Naito Y, Andoh A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin J Gastroenterol 2018 Feb;11(1):1-10.
  23. Scher JU, Littman DR, Abramson SB. Review: Microbiome in Inflammatory Arthritis and Human Rheumatic Diseases. Arthritis Rheumatol 2016 Jan;68(1):35-45.
  24. Asquith MJ, Stauffer P, Davin S, Mitchell C, Lin P, Rosenbaum JT. Perturbed Mucosal Immunity and Dysbiosis Accompany Clinical Disease in a Rat Model of Spondyloarthritis. Arthritis Rheumatol (Hoboken, NJ) 2016;68(9):2151-62.
  25. Gill T, Asquith M, Brooks SR, Rosenbaum JT, Colbert RA. Effects of HLA-B27 on Gut Microbiota in Experimental Spondyloarthritis Implicate an Ecological Model of Dysbiosis. Arthritis Rheumatol 2018;70(4):555-65.
  26. Asquith M, Sternes PR, Costello ME, Karstens L, Diamond S, Martin TM, et al. HLA Alleles Associated With Risk of Ankylosing Spondylitis and Rheumatoid Arthritis Influence the Gut Microbiome. Arthritis Rheumatol 2019;71(10):1642-50.
  27. Costello M-E, Ciccia F, Willner D, Warrington N, Robinson PC, Gardiner B, et al. Brief Report: Intestinal Dysbiosis in Ankylosing Spondylitis. Arthritis Rheumatol 2015 Mar;67(3):686-91.
  28. Ciccia F, Accardo-Palumbo A, Rizzo A, Guggino G, Raimondo S, Giardina A, et al. Evidence that autophagy, but not the unfolded protein response, regulates the expression of IL-23 in the gut of patients with ankylosing spondylitis and subclinical gut inflammation. Ann Rheum Dis 2014 Aug 5;73(8):1566-74.
  29. Angelidou I, Chrysanthopoulou A, Mitsios A, Arelaki S, Arampatzioglou A, Kambas K, et al. REDD1/Autophagy Pathway Is Associated with Neutrophil-Driven IL-1β Inflammatory Response in Active Ulcerative Colitis. J Immunol 2018 Jun 15;200(12):3950-61.
  30. Ciccia F, Guggino G, Rizzo A, Saieva L, Peralta S, Giardina A, et al. Type 3 innate lymphoid cells producing IL-17 and IL-22 are expanded in the gut, in the peripheral blood, synovial fluid and bone marrow of patients with ankylosing spondylitis. Ann Rheum Dis 2015 Sep;74(9):1739-47.
  31. Ciccia F, Bombardieri M, Principato A, Giardina A, Tripodo C, Porcasi R, et al. Overexpression of interleukin-23, but not interleukin-17, as an immunologic signature of subclinical intestinal inflammation in ankylosing spondylitis. Arthritis Rheum 2009;60(4):955-65.
  32. Sherlock JP, Joyce-Shaikh B, Turner SP, Chao C, Sathe M, Grein J, et al. IL-23 induces spondyloarthropathy by acting on ROR-γt+ CD3+CD4-CD8- entheseal resident T cells. Nat Med 2012 Jul;18(7):1069-76.
  33. Cuthbert RJ, Fragkakis EM, Dunsmuir R, Li Z, Coles M, Marzo-Ortega H, et al. Group 3 Innate Lymphoid Cells in Human Enthesis. Arthritis Rheumatol 2017;69(9):1816-22.
  34. Cuthbert RJ, Watad A, Fragkakis EM, Dunsmuir R, Loughenbury P, Khan A, et al. Evidence that tissue resident human enthesis γδT-cells can produce IL-17A independently of IL-23R transcript expression. Ann Rheum Dis 2019 Nov 17;78(11):1559-65.
  35. Di Carlo M, Luchetti MM, Benfaremo D, Di Donato E, Mosca P, Maltoni S, et al. The DETection of Arthritis in Inflammatory boweL diseases (DETAIL) questionnaire: development and preliminary testing of a new tool to screen patients with inflammatory bowel disease for the presence of spondyloarthritis. Clin Rheumatol 2018;37(4):1037-44.
  36. Variola A, Zanolin ME, Cipriano G, Macchioni P, Martinis F, Pasetti A, et al. The IBIS-Q [IBd Identification of Spondyloarthritis Questionnaire]: A Novel Tool to Detect Both Axial and Peripheral Arthritis in Inflammatory Bowel Disease Patients. J Crohn’s Colitis 2020;14(12):1680-6.
  37. Hasler S, Zahnd N, Müller S, Vavricka S, Rogler G, Tandjung R, et al. VAlidation of an 8-item-questionnaire predictive for a positive caLprotectin tEst and Real-life implemenTation in primary care to reduce diagnostic delay in inflammatory bowel disease (ALERT): protocol for a prospective diagnostic study. BMJ Open 2015 Mar 10;5(3):e007306.
  38. Sanz Sanz J, Juanola Roura X, Seoane-Mato D, Montoro M, Gomollón F, Grupo de Trabajo del proyecto PIIASER. Screening of Inflammatory Bowel Disease and Spondyloarthritis for Referring Patients Between Rheumatology and Gastroenterology. Reumatol Clin 2018;14(2):68-74.
  39. Felice C, Leccese P, Scudeller L, Lubrano E, Cantini F, Castiglione F, et al. Red flags for appropriate referral to the gastroenterologist and the rheumatologist of patients with inflammatory bowel disease and spondyloarthritis. Clin Exp Immunol 2019 Apr 9;196(1):123-38.
  40. Sieper J, van der Heijde D, Landewé R, Brandt J, Burgos-Vagas R, Collantes-Estevez E, et al. New criteria for inflammatory back pain in patients with chronic back pain: a real patient exercise by experts from the Assessment of SpondyloArthritis international Society (ASAS). Ann Rheum Dis 2009 Jun;68(6):784-8.
  41. Gionchetti P, Calabrese C, Rizzello F. Inflammatory bowel diseases and spondyloarthropathies. J Rheumatol 2015 Nov 1;93:21-3.
  42. Olivieri I, Cantini F, Castiglione F, Felice C, Gionchetti P, Orlando A, et al. Italian Expert Panel on the management of patients with coexisting spondyloarthritis and inflammatory bowel disease. Autoimmun Rev 2014 Aug;13(8):822-30.
  43. Danese S, Fiorino G, Mary J-Y, Lakatos PL, Haens GD’, Moja L, et al. Development of Red Flags Index for Early Referral of Adults with Symptoms and Signs Suggestive of Crohn’s Disease: An IOIBD Initiative. J Crohn’s Colitis 2015;601-6.
  44. Rudwaleit M, Khan M a, Sieper J. The challenge of diagnosis and classification in early ankylosing spondylitis: do we need new criteria? Arthritis Rheum 2005 Apr;52(4):1000-8.
  45. Benhamou M, Gossec L, Dougados M. Clinical relevance of C-reactive protein in ankylosing spondylitis and evaluation of the NSAIDs/coxibs’ treatment effect on C-reactive protein. Rheumatology 2009;49(3):536-41.
  46. Harbord M, Annese V, Vavricka SR, Allez M, Acosta MB de, Boberg KM, et al. The first european evidence-based consensus on extra-intestinal manifestations in inflammatory bowel disease. J Crohn’s Colitis 2016;10(3):239-54.
  47. Steer S, Jones H, Hibbert J, Kondeatis E, Vaughan R, Sanderson J, et al. Low back pain, sacroiliitis, and the relationship with HLA-B27 in Crohn’s disease. J Rheumatol 2003 Mar;30(3):518-22.
  48. Palm O, Moum B, Ongre A, Gran JT. Prevalence of ankylosing spondylitis and other spondyloarthropathies among patients with inflammatory bowel disease: a population study (the IBSEN study). J Rheumatol 2002 Mar;29(3):511-5.
  49. Østgård RD, Deleuran BW, Dam MY, Hansen IT, Jurik AG, Glerup H. Faecal calprotectin detects subclinical bowel inflammation and may predict treatment response in spondyloarthritis. Scand J Rheumatol 2018 Jan 2;47(1):48-55.
  50. Klingberg E, Carlsten H, Hilme E, Hedberg M, Forsblad-D’Elia H. Calprotectin in ankylosing spondylitis - Frequently elevated in feces, but normal in serum. Scand J Gastroenterol 2012 Apr;47(4):435-44.
  51. Cypers H, Varkas G, Beeckman S, Debusschere K, Vogl T, Roth J, et al. Elevated calprotectin levels reveal bowel inflammation in spondyloarthritis. Ann Rheum Dis 2016 Jul;75(7):1357-62.
  52. Fauny M, D’Amico F, Bonovas S, Netter P, Danese S, Loeuille D, et al. Faecal Calprotectin for the Diagnosis of Bowel Inflammation in Patients With Rheumatological Diseases: A Systematic Review. J Crohns Colitis 2020 Jun 19;14(5):688-93.
  53. O’Brien J. Nonsteroidal anti-inflammatory drugs in patients with inflammatory bowel disease. Am J Gastroenterol 2000 Aug;95(8):1859-61.
  54. van der Heijde D, Ramiro S, Landewé R, Baraliakos X, Van den Bosch F, Sepriano A, et al. 2016 update of the ASAS-EULAR management recommendations for axial spondyloarthritis. Ann Rheum Dis 2017 Jun;76(6):978-91.
  55. Ward MM, Deodhar A, Gensler LS, Dubreuil M, Yu D, Khan MA, et al. 2019 Update of the American College of Rheumatology / Spondylitis Association of America / Spondyloarthritis Research and Treatment Network Recommendations for the Treatment of Ankylosing Spondylitis and Nonradiographic Axial Spondyloarthritis 2019;71(10):1285-99.
  56. Ananthakrishnan AN, Higuchi LM, Huang ES, Khalili H, Richter JM, Fuchs CS, et al. Aspirin, nonsteroidal anti-inflammatory drug use, and risk for crohn disease and ulcerative colitis. Ann Intern Med 2012 Mar 6;156(5):350-9.
  57. Felder JB, Korelitz BI, Rajapakse R, Schwarz S, Horatagis AP, Gleim G. Effects of nonsteroidal antiinflammatory drugs on inflammatory bowel disease: a case-control study. Am J Gastroenterol 2000 Aug;95(8):1949-54.
  58. Bonner GF, Fakhri A, Vennamanemi SR. A long-term cohort study of nonsteroidal anti-inflammatory drug use and disease activity in outpatients with inflammatory bowel disease. Inflamm Bowel Dis 2004 Nov;10(6):751-7.
  59. El Miedany Y, Youssef S, Ahmed I, El Gaafary M. The gastrointestinal safety and effect on disease activity of etoricoxib, a selective Cox-2 inhibitor in inflammatory bowel diseases. Am J Gastroenterol 2006 Feb;101(2):311-7.
  60. Sandborn WJ, Stenson WF, Brynskov J, Lorenz RG, Steidle GM, Robbins JL, et al. Safety of celecoxib in patients with ulcerative colitis in remission: A randomized, placebo-controlled, pilot study. Clin Gastroenterol Hepatol 2006 Feb;4(2):203-11.
  61. Ribaldone DG, Fagoonee S, Astegiano M, De Angelis C, Smedile A, Caviglia GP, et al. Coxib’s Safety in Patients with Inflammatory Bowel Diseases: A Meta-analysis. Pain Physician 2015 Nov;18(6):599-607.
  62. Miao X-P, Li J-S, Ouyang Q, Hu R-W, Zhang Y, Li H-Y. Tolerability of selective cyclooxygenase 2 inhibitors used for the treatment of rheumatological manifestations of inflammatory bowel disease. Cochrane Database Syst Rev 2014 Oct 23;2014(10):CD007744.
  63. Summers RW, Switz DM, Sessions JT, Becktel JM, Best WR, Kern F, et al. National Cooperative Crohn’s Disease Study: results of drug treatment. Gastroenterology 1979 Oct;77(4 Pt 2):847-69.
  64. Malchow H, Ewe K, Brandes JW, Goebell H, Ehms H, Sommer H, et al. European Cooperative Crohn’s Disease Study (ECCDS): results of drug treatment. Gastroenterology 1984 Feb;86(2):249-66.
  65. Gomollón F, Dignass A, Annese V, Tilg H, Van Assche G, Lindsay JO, et al. 3rd European evidence-based consensus on the diagnosis and management of Crohn’s disease 2016: Part 1: Diagnosis and medical management. J Crohn’s Colitis 2017 Jan 1;11(1):3-25.
  66. Haibel H, Fendler C, Listing J, Callhoff J, Braun J, Sieper J. Efficacy of oral prednisolone in active ankylosing spondylitis: results of a double-blind, randomised, placebo-controlled short-term trial. Ann Rheum Dis 2014 Jan 26;73(1):243-6.
  67. Gossec L, Baraliakos X, Kerschbaumer A, de Wit M, McInnes I, Dougados M, et al. EULAR recommendations for the management of psoriatic arthritis with pharmacological therapies: 2019 update. Ann Rheum Dis 2020 Jun 20;79(6):700-12.
  68. Bergman R, Parkes M. Systematic review: the use of mesalazine in inflammatory bowel disease. Aliment Pharmacol Ther 2006 Apr;23(7):841-55.
  69. Murray A, Nguyen TM, Parker CE, Feagan BG, MacDonald JK. Oral 5-aminosalicylic acid for induction of remission in ulcerative colitis. Cochrane Database Syst Rev 2020 Aug 12;8(8):CD000543.
  70. Dougados M, Linden S VAN DER, Leirisalo‐Repo M, Huitfeldt B, Juhlin R, Veys E, et al. Sulfasalazine in the treatment of spondylarthropathy. Arthritis Rheum 1995;38(5):618-27.
  71. Clegg DO, Reda DJ, Abdellatif M. Comparison of sulfasalazine and placebo for the treatment of axial and peripheral articular manifestations of the seronegative spondylarthropathies: a Department of Veterans Affairs cooperative study. Arthritis Rheum 1999 Nov;42(11):2325-9.
  72. Braun J, Zochling J, Baraliakos X, Alten R, Burmester G, Grasedyck K, et al. Efficacy of sulfasalazine in patients with inflammatory back pain due to undifferentiated spondyloarthritis and early ankylosing spondylitis: A multicentre randomised controlled trial. Ann Rheum Dis 2006 Sep;65(9):1147-53.
  73. Chen J, Lin S, Liu C. Sulfasalazine for ankylosing spondylitis. Cochrane Database Syst Rev 2014 Nov 27;2014(11):CD004800.
  74. Herfarth HH. Methotrexate for Inflammatory Bowel Diseases - New Developments. J Dig Dis 2016;34(1-2):140-6.
  75. Herfarth H, Barnes EL, Valentine JF, Hanson J, Higgins PDR, Isaacs KL, et al. Methotrexate Is Not Superior to Placebo in Maintaining Steroid-Free Response or Remission in Ulcerative Colitis. Gastroenterology 2018 Oct;155(4):1098-1108.e9.
  76. Carbonnel F, Colombel JF, Filippi J, Katsanos KH, Peyrin-Biroulet L, Allez M, et al. Methotrexate Is Not Superior to Placebo for Inducing Steroid-Free Remission, but Induces Steroid-Free Clinical Remission in a Larger Proportion of Patients With Ulcerative Colitis. Gastroenterology 2016 Feb;150(2):380-388.e4.
  77. Chen J, Veras MMS, Liu C, Lin J. Methotrexate for ankylosing spondylitis. Cochrane Database Syst Rev 2013 Feb 28;2013(2):CD004524.
  78. Haibel H, Brandt HC, Song IH, Brandt A, Listing J, Rudwaleit M, et al. No efficacy of subcutaneous methotrexate in active ankylosing spondylitis: a 16-week open-label trial. Ann Rheum Dis 2007 Mar 1;66(3):419-21.
  79. Kaltwasser JP, Nash P, Gladman D, Rosen CF, Behrens F, Jones P, 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 Jun;50(6):1939-50.
  80. van Denderen JC, van der Paardt M, Nurmohamed MT, de Ryck YMMA, Dijkmans BAC, van der Horst-Bruinsma IE. Double blind, randomised, placebo controlled study of leflunomide in the treatment of active ankylosing spondylitis. Ann Rheum Dis 2005 Dec;64(12):1761-4.
  81. Prajapati DN, Knox JF, Emmons J, Saeian K, Csuka ME, Binion DG. Leflunomide treatment of Crohn’s disease patients intolerant to standard immunomodulator therapy. J Clin Gastroenterol 2003 Aug;37(2):125-8.
  82. Holtmann MH, Gerts AL, Weinman A, Galle PR, Neurath MF. Treatment of Crohn’s disease with leflunomide as second-line immunosuppression: A phase 1 open-label trial on efficacy, tolerability and safety. Dig Dis Sci 2008 Apr;53(4):1025-32.
  83. Timmer A, Patton PH, Chande N, McDonald JWD, MacDonald JK. Azathioprine and 6-mercaptopurine for maintenance of remission in ulcerative colitis. Cochrane Database Syst Rev 2016 May 18;2016(5):CD000478.
  84. Chande N, Patton PH, Tsoulis DJ, Thomas BS, MacDonald JK. Azathioprine or 6-mercaptopurine for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev 2015 Oct 30;2017(10):CD000067.
  85. Generini S, Giacomelli R, Fedi R, Fulminis A, Pignone A, Frieri G, et al. Infliximab in spondyloarthropathy associated with Crohn’s disease: An open study on the efficacy of inducing and maintaining remission of musculoskeletal and gut manifestations. Ann Rheum Dis 2004 Dec;63(12):1664-9.
  86. Conigliaro P, Chimenti MS, Triggianese P, D’Antonio A, Sena G, Alfieri N, et al. Two years follow-up of golimumab treatment in refractory enteropathic spondyloarthritis patients with Crohn disease: A STROBE-compliant study. Medicine (Baltimore) 2021 Mar 26;100(12):e25122.
  87. Marzo-Ortega H, McGonagle D, O’Connor P, Emery P. Efficacy of etanercept for treatment of Crohn’s related spondyloarthritis but not colitis. Ann Rheum Dis 2003 Jan 1;62(1):74-6.
  88. van der Heijde D, Dijkmans B, Geusens P, Sieper J, DeWoody K, Williamson P, et al. Efficacy and safety of infliximab in patients with ankylosing spondylitis: Results of a randomized, placebo-controlled trial (ASSERT). Arthritis Rheum 2005 Feb;52(2):582-91.
  89. Davis JC, van der Heijde DM, Braun J, Dougados M, Cush J, Clegg D, et al. Sustained durability and tolerability of etanercept in ankylosing spondylitis for 96 weeks. Ann Rheum Dis 2005 Nov;64(11):1557-62.
  90. van der Heijde D, Kivitz A, Schiff MH, Sieper J, Dijkmans BAC, Braun J, et al. Efficacy and safety of adalimumab in patients with ankylosing spondylitis: results of a multicenter, randomized, double-blind, placebo-controlled trial. Arthritis Rheum 2006 Jul;54(7):2136-46.
  91. Inman RD, Davis JC, Van Der Heijde D, Diekman L, Sieper J, Kim S Il, et al. Efficacy and safety of golimumab in patients with ankylosing spondylitis: results of a randomized, double-blind, placebo-controlled, phase III trial. Arthritis Rheum 2008 Nov;58(11):3402-12.
  92. Landewé R, Braun J, Deodhar A, Dougados M, Maksymowych WP, Mease PJ, et al. Efficacy of certolizumab pegol on signs and symptoms of axial spondyloarthritis including ankylosing spondylitis: 24-week results of a double-blind randomised placebo-controlled Phase 3 study. Ann Rheum Dis 2014;73:39-47.
  93. Baeten D, Sieper J, Braun J, Baraliakos X, Dougados M, Emery P, et al. Secukinumab, an Interleukin-17A Inhibitor, in Ankylosing Spondylitis. N Engl J Med 2015;373(26):2534-48.
  94. van der Heijde D, Cheng-Chung Wei J, Dougados M, Mease P, Deodhar A, Maksymowych WP, et al. Ixekizumab, an interleukin-17A antagonist in the treatment of ankylosing spondylitis or radiographic axial spondyloarthritis in patients previously untreated with biological disease-modifying anti-rheumatic drugs (COAST-V): 16 week results of a phase 3 randomised, double-blind, active-controlled and placebo-controlled trial. Lancet 2018 Dec 8;392(10163):2441-51.
  95. Carron P, Varkas G, Cypers H, Van Praet L, Elewaut D, Van den Bosch F. Anti-TNF-induced remission in very early peripheral spondyloarthritis: the CRESPA study. Ann Rheum Dis 2017 Aug;76(8):1389-95.
  96. Mease P, Sieper J, Van den Bosch F, Rahman P, Karunaratne PM, Pangan AL. Randomized Controlled Trial of Adalimumab in Patients With Nonpsoriatic Peripheral Spondyloarthritis. Arthritis Rheumatol 2015;67(4):914-23.
  97. Paramarta JE, De Rycke L, Heijda TF, Ambarus CA, Vos K, Dinant HJ, et al. Efficacy and safety of adalimumab for the treatment of peripheral arthritis in spondyloarthritis patients without ankylosing spondylitis or psoriatic arthritis. Ann Rheum Dis 2013 Nov 8;72(11):1793-9.
  98. Marzo-Ortega H, McGonagle D, O’Connor P, Emery P. Efficacy of Etanercept in the Treatment of the Entheseal Pathology in Resistant Spondylarthropathy: A Clinical and Magnetic Resonance Imaging Study. Arthritis Rheum 2001;44(9):2112-7.
  99. Hanauer SB, Feagan BG, Lichtenstein GR, Mayer LF, Schreiber S, Colombel JF, et al. Maintenance infliximab for Crohn’s disease: The ACCENT I randomised trial. Lancet 2002 May 4;359(9317):1541-9.
  100. Rutgeerts P, Sandborn WJ, Feagan BG, Reinisch W, Olson A, Johanns J, et al. Infliximab for Induction and Maintenance Therapy for Ulcerative Colitis. N Engl J Med 2005 Dec 8;353(23):2462-76.
  101. Hanauer SB, Sandborn WJ, Rutgeerts P, Fedorak RN, Lukas M, Macintosh D, et al. Human anti-tumor necrosis factor monoclonal antibody (adalimumab) in Crohn’s disease: The CLASSIC-I trial. Gastroenterology 2006;130(2):323-33.
  102. Reinisch W, Sandborn WJ, Hommes DW, D’Haens G, Hanauer S, Schreiber S, et al. Adalimumab for induction of clinical remission in moderately to severely active ulcerative colitis: Results of a randomised controlled trial. Gut 2011 Jun;60(6):780-7.
  103. Sandborn WJ, Feagan BG, Stoinov S, Honiball PJ, Rutgeerts P, Mason D, et al. Certolizumab Pegol for the Treatment of Crohn’s Disease. N Engl J Med 2007 Jul 19;357(3):228-38.
  104. Sandborn WJ, Feagan BG, Marano C, Zhang H, Strauss R, Johanns J, et al. Subcutaneous golimumab induces clinical response and remission in patients with moderate-to-severe ulcerative colitis. Gastroenterology 2014;146(1):85-95.
  105. Feagan BG, Sandborn WJ, Gasink C, Jacobstein D, Lang Y, Friedman JR, et al. Ustekinumab as Induction and Maintenance Therapy for Crohn’s Disease. N Engl J Med 2016 Nov 17;375(20):1946-60.
  106. Sandborn WJ, Feagan BG, Rutgeerts P, Hanauer S, Colombel J-F, Sands BE, et al. Vedolizumab as Induction and Maintenance Therapy for Crohn’s Disease. N Engl J Med 2013 Aug 22;369(8):711-21.
  107. Feagan BG, Rutgeerts P, Sands BE, Hanauer S, Colombel J-F, Sandborn WJ, et al. Vedolizumab as Induction and Maintenance Therapy for Ulcerative Colitis. N Engl J Med 2013 Aug 22;369(8):699-710.
  108. Sands BE, Sandborn WJ, Panaccione R, O’Brien CD, Zhang H, Johanns J, et al. Ustekinumab as Induction and Maintenance Therapy for Ulcerative Colitis. N Engl J Med 2019 Sep 26;381(13):1201-14.
  109. McInnes IB, Kavanaugh A, Gottlieb AB, Puig L, Rahman P, Ritchlin C, et al. Efficacy and safety of ustekinumab in patients with active psoriatic arthritis: 1 year results of the phase 3, multicentre, double-blind, placebo-controlled PSUMMIT 1 trial. Lancet 2013 Aug 31;382(9894):780-9.
  110. Ritchlin C, Rahman P, Kavanaugh A, McInnes IB, Puig L, Li S, et al. Efficacy and safety of the anti-IL-12/23 p40 monoclonal antibody, ustekinumab, in patients with active psoriatic arthritis despite conventional non-biological and biological anti-tumour necrosis factor therapy: 6-month and 1-year results of the phase 3, m. Ann Rheum Dis 2014 Jun 1;73(6):990-9.
  111. Pugliese D, Daperno M, Fiorino G, Savarino E, Mosso E, Biancone L, et al. Real-life effectiveness of ustekinumab in inflammatory bowel disease patients with concomitant psoriasis or psoriatic arthritis: An IG-IBD study. Dig Liver Dis 2019 Jul 1;51(7):972-7.
  112. Deodhar A, Gensler LS, Sieper J, Clark M, Calderon C, Wang Y, et al. Three Multicenter, Randomized, Double-Blind, Placebo-Controlled Studies Evaluating the Efficacy and Safety of Ustekinumab in Axial Spondyloarthritis. Arthritis Rheumatol (Hoboken, NJ) 2019 Feb;71(2):258-70.
  113. McInnes IB, Mease PJ, Kirkham B, Kavanaugh A, Ritchlin CT, Rahman P, et al. Secukinumab, a human anti-interleukin-17A monoclonal antibody, in patients with psoriatic arthritis (FUTURE 2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2015 Sep;386(9999):1137-46.
  114. Deodhar A, Blanco R, Dokoupilová E, Hall S, Kameda H, Kivitz AJ, et al. Improvement of Signs and Symptoms of Nonradiographic Axial Spondyloarthritis in Patients Treated With Secukinumab: Primary Results of a Randomized, Placebo‐Controlled Phase III Study. Arthritis Rheumatol 2021 Jan 24;73(1):110-20.
  115. Hueber W, Sands BE, Lewitzky S, Vandemeulebroecke M, Reinisch W, Higgins PDR, et al. Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn’s disease: unexpected results of a randomised, double-blind placebo-controlled trial. Gut 2012 Dec;61(12):1693-700.
  116. Fauny M, Moulin D, D’Amico F, Netter P, Petitpain N, Arnone D, et al. Paradoxical gastrointestinal effects of interleukin-17 blockers. Ann Rheum Dis 2020 Sep;79(9):1132-8.
  117. Schreiber S, Colombel J-F, Feagan BG, Reich K, Deodhar AA, McInnes IB, et al. Incidence rates of inflammatory bowel disease in patients with psoriasis, psoriatic arthritis and ankylosing spondylitis treated with secukinumab: a retrospective analysis of pooled data from 21 clinical trials. Ann Rheum Dis 2019;78(4):473-9.
  118. Burisch J, Eigner W, Schreiber S, Aletaha D, Weninger W, Trauner M, et al. Risk for development of inflammatory bowel disease under inhibition of interleukin 17: A systematic review and meta-analysis. PLoS One 2020;15(5):e0233781.
  119. Feagan BG, Sandborn WJ, Colombel J-F, Byrne SO, Khalid JM, Kempf C, et al. Incidence of Arthritis/Arthralgia in Inflammatory Bowel Disease with Long-term Vedolizumab Treatment: Post Hoc Analyses of the GEMINI Trials. J Crohns Colitis 2019 Jan 1;13(1):50-7.
  120. Dubash S, Marianayagam T, Tinazzi I, Al-Araimi T, Pagnoux C, Weizman A V., et al. Emergence of severe spondyloarthropathy-related entheseal pathology following successful vedolizumab therapy for inflammatory bowel disease. Rheumatol (United Kingdom) 2019;58(6):963-8.
  121. Varkas G, Thevissen K, De Brabanter G, Van Praet L, Czul-gurdian F, Cypers H, et al. An induction or flare of arthritis and/or sacroiliitis by vedolizumab in inflammatory bowel disease: a case series. Ann Rheum Dis 2017 May;76(5):878-81.
  122. Tadbiri S, Peyrin-Biroulet L, Serrero M, Filippi J, Pariente B, Roblin X, et al. Impact of vedolizumab therapy on extra-intestinal manifestations in patients with inflammatory bowel disease: a multicentre cohort study nested in the OBSERV-IBD cohort. Aliment Pharmacol Ther 2018;47(4):485-93.
  123. Sandborn WJ, Ghosh S, Panes J, Vranic I, Su C, Rousell S, et al. Tofacitinib, an Oral Janus Kinase Inhibitor, in Active Ulcerative Colitis. N Engl J Med 2012 Aug 16;367(7):616-24.
  124. Sandborn WJ, Su C, Sands BE, D’Haens GR, Vermeire S, Schreiber S, et al. Tofacitinib as Induction and Maintenance Therapy for Ulcerative Colitis. N Engl J Med 2017 May 4;376(18):1723-36.
  125. Mease P, Hall S, FitzGerald O, van der Heijde D, Merola JF, Avila-Zapata F, et al. Tofacitinib or Adalimumab versus Placebo for Psoriatic Arthritis. N Engl J Med 2017;377(16):1537-50.
  126. Gladman D, Rigby W, Azevedo VF, Behrens F, Blanco R, Kaszuba A, et al. Tofacitinib for Psoriatic Arthritis in Patients with an Inadequate Response to TNF Inhibitors. N Engl J Med 2017;377(16):1525-36.
  127. Deodhar A, Sliwinska-Stanczyk P, Xu H, Baraliakos X, Gensler LS, Fleishaker D, et al. Tofacitinib for the treatment of ankylosing spondylitis: a phase III, randomised, double-blind, placebo-controlled study. Ann Rheum Dis 2021 Apr 27;annrheumdis-2020-219601.
  128. Wang W, Cleveland NK, Ollech J, Rubin DT. Use of Tofacitinib for the Treatment of Arthritis Associated With Ulcerative Colitis. ACG Case Reports J 2019 Sep;6(9):e00226.
  129. Nwaogu A, Bond A, Smith PJ. Guideline review: Tofacitinib for adults with moderately to severely active ulcerative colitis - NICE guidance. Frontline Gastroenterol 2021 Mar 1;12(2):133-6.
  130. Lee JA, Magavi PR, Konijeti GG. Treatment of Ulcerative Colitis and Seronegative Inflammatory Spondyloarthritis With Vedolizumab and Tofacitinib. Inflamm Bowel Dis 2020 Nov 23;26(11):e146.
  131. Le Berre C, Loeuille D, Peyrin-Biroulet L. Combination Therapy With Vedolizumab and Tofacitinib in a Patient With Ulcerative Colitis and Spondyloarthropathy. Clin Gastroenterol Hepatol 2019 Mar 1;17(4):794-6.
  132. Roblin X, Paul S, Ben-Horin S. Co-treatment With Golimumab and Vedolizumab to Treat Severe UC and Associated Spondyloarthropathy. J Crohns Colitis 2018 Feb 28;12(3):379-80.
  133. Bethge J, Meffert S, Ellrichmann M, Conrad C, Nikolaus S, Schreiber S. Combination therapy with vedolizumab and etanercept in a patient with pouchitis and spondylarthritis. BMJ Open Gastroenterol 2017 Feb 1;4(1):e000127.
  134. Privitera G, Pugliese D, Onali S, Petito V, Scaldaferri F, Gasbarrini A, et al. Combination therapy in inflammatory bowel disease - from traditional immunosuppressors towards the new paradigm of dual targeted therapy. Autoimmun Rev 2021 Jun;20(6):102832.