Printer Friendly

Update on the Medical Management of Gastrointestinal Behcet's Disease.

1. Introduction

Behcet's disease (BD) is a rare relapsing systemic inflammatory disorder of unknown etiology characterized by recurrent oral ulcers, genital sores, and ocular lesions; however many other organs including the vascular, neurological, and musculoskeletal systems as well as the gastrointestinal system can be involved [1-3]. Genetic and environmental factors play a key role in this disorder, in particular the human leukocyte antigen B51 allele, located in the major histocompatibility complex locus, representing the strongest risk factor for the development of BD [4]. In recent years, some microbial agents such as Herpes simplex virus 1 and Streptococcus sanguinis have gained increasing importance as potential infectious agents of BD [5], being able to generate an inflammatory process leading to a CD4+ T lymphocytes clonal expansion which in turn produces high concentrations of both proinflammatory cytokines and cytotoxic CD8+ cells [6]. Several cytokines are claimed to contribute to the pathological scenario of BD [5, 7-9]: tumor necrosis factor- (TNF-) [alpha] partakes probably in somehow the disease onset and the successful use of anti-TNF-[alpha] agents has substantiated the role of this cytokine in BD [10-12]. Conversely interleukin- (IL-) 6 seems to be related to central nervous system involvement, as confirmed by its high levels in the cerebral spinal fluid of affected patients [13]. Recent studies have also suggested a role of IL-1, since its secretion in BD patients appeared to be related to NLRP3 inflammasome activation [14-17].

Although oral aphthae and genital ulcers are the earliest and the most frequent manifestations of BD, anticipating by many years other typical BD clinical symptoms, GIBD is one of the major causes of morbidity and mortality, often leading to severe complications. GIBD occurs in 3-60% of patients, on average 4.5-6 years after the onset of oral ulcerations [18], varying among different populations [2,19-21] and being more frequent in Japan, United Kingdom, and Taiwan than in the Middle East and Mediterranean basin [4, 22, 23]. Intestinal ulcerations are the main pathological features of GIBD, and it is thought they are secondary to small vessel vasculitis, albeit a large vessel involvement leading to ischemic damage may arise [24]. GIBD may be suspected when diarrhoea, melena, and hematochezia occur [25-27]. Common complaints may include abdominal pain, fever, anorexia, vomiting, and dyspepsia, and a palpable mass on the affected quadrant can also be noticed [25]. The terminal ileum is the most common localization of disease, followed by the ileocecal region and colon [28]; esophagus engagement is unusual [29], and rectum or anus is also rarely affected [30] while the stomach is the least frequently involved part of the gastrointestinal tract [31,32]. Table 1 shows the most common intestinal localizations of BD.

Gastrointestinal lesions are typically irregular, round or oval, punched-out, large (>1 cm), single to a few in number, deep, and with discrete margins in a focal distribution [33]. On the basis of endoscopic findings, they are classified into volcano, geographic, and aphthous types. The volcano-type, deeply penetrating and having nodular margins caused by fibrosis, is strictly associated with a poor prognosis [34, 35]. The differential diagnosis between GIBD and inflammatory bowel disease, in particular Crohn's disease (CD), is often difficult, albeit in the latter the ulcers have typically a cobblestone appearance with a segmental distribution which involves irregularly different parts of the gastrointestinal tract [33]. In this regard a diagnostic algorithm using a classification analysis of the lesions has been proposed in order to identify valuable strategies for differential diagnosis [36]. A clinical scoring system known as the Disease Activity Index for Intestinal BD (DAIBD) provides a score between 0 and 325 based on an 8-point index; it classifies disease activity as quiescent ([less than or equal to]19), mild (20-39), moderate (40- 74), and severe ([greater than or equal to]75) on the basis of patient's general condition, extraintestinal manifestations, intestinal symptoms and signs, and stool frequency [37]. Table 1 summarizes the main clinical, endoscopic, and pathological findings of GIBD.

Although a wide number of conventional immunosuppressive drugs have been used to induce remission in GIBD, several failures have been reported. This article reviews the progress in the management of GIBD focusing on current treatment strategies and possible future perspectives. An electronic literature search was conducted using the PubMed database and the clinicaltrials.gov search engine. We looked for all studies published in the last years, including case reports, clinical trials, and cohort studies (Table 2).

2. The Management of GIBD

As underlined in the guidelines of the European League Against Rheumatism (EULAR) for the management of BD, evidence-based recommendations regarding GIBD are not provided due to the poor amount of published clinical trials [38]. Medical treatment such as corticosteroids (CC), sulfasalazine (SSZ), and azathioprine (AZA) are capable of inducing remission without the need for surgery in many patients [25, 39], whereas TNF-[alpha] antagonists and thalidomide (THD) have proven useful in resistant and complicated cases [10-12, 40, 41].

Nevertheless GIBD management is still largely empirical due to the lack of not yet standardized medical treatments, the heterogeneity of this disorder, and the unpredictable exacerbations of BD. To date, several conventional immunosuppressive drugs may be employed, although none of them has been proven actually effective in preventing disease relapse.

2.1. Corticosteroids (CC). CC are the first-line therapy, especially in patients with severe systemic symptoms, recurrent gastrointestinal bleeding, or when treatment with 5aminosalicylic acid (5-ASA)/SSZ is not enough [42]. CC are supposed to be very effective in the short term; indeed it is widely accepted to start with prednisolone or its equivalent at 0.5-1 mg/kg which has to be quickly tapered by 5 mg each week within few months [43]. The rationale behind this strategy may be figured out noticing the evidence reported by Park et al. in a retrospective cohort study; systemic CC therapy (mean starting dose, 0.58 mg/kg) was administered in 54 patients with active GIBD; a complete remission and partial remission were achieved in 46.3% and 42.6% of patients respectively, whereas only 11.1% showed no response one month after starting treatment. At one-year follow-up, a prolonged response was found in 26 out of 54 patients, whereas 19 patients showed CC dependency, suggesting that their employment is not desirable over extended periods [44]. Several literature data report the association between CC employment and GI side effects, including bleeding or perforation. GI bleeding and perforation are assumed to occur when ulcers erode into underlying vessels. CC may impair tissue repair, thus leading to delayed wound healing. Despite these assumptions, a recent systematic review and meta-analysis of randomised, double-blind, controlled trials comparing CC to placebo for any medical condition or in healthy participants have suggested that additional factors to corticosteroid therapy, such as severe physiological stress, may decrease mucosal blood flow with subsequent tissue ischemia making some patients more vulnerable to adverse events under CC assumption. Therefore acid-suppressive therapy may be considered a valuable aid in preventing the occurrence of ulcers in clinical settings [45].

2.2. 5-Aminosalicylic Acid (5-ASA)/Sulfasalazine (SSZ). 5-ASA/SSZ is indicated in all cases of GIBD due to its safety profile and current limited therapeutic options [46]. It is usually administered at a dose of 2-4 g/day for inducing remission in mild forms of BD and for maintenance once remission is achieved [27]. Convincing evidence about the efficacy of 5-ASA derives from nonrandomized studies and case series suggesting that it is effective in treating esophageal and gastrointestinal manifestations of BD [47, 48], albeit conflicting data regarding its usefulness have been reported as well [49]. In a retrospective cohort study investigating 143 patients with GIBD who received 5-ASA/SSZ alone for maintaining remission, cumulative relapse rates at 1, 3, 5, and 10 years after remission were 8.1%, 22.6%, 31.2%, and 46.7%, respectively. Of note, a younger age at diagnosis (<35 years), higher serum level of C-reactive protein (1.5 mg/dL), and greater DAIBD score ([greater than or equal to]60) were regarded as independent predictors of relapse [50]. More recently an observational study has suggested 5-ASA as a valuable treatment for preventing postoperative recurrences; remission was achieved in 10 out of 16 (62.5%) patients who took 5-ASA compounds, and no exacerbation was seen during the 89.3 [+ or -] 64.5 months that they were followed. Similarly, remission was observed also among 37 patients who were prescribed with azathioprine, and there were no relapses in 24/37 (65%) patients during a mean follow-up of 68.6 [+ or -] 43.6 months [51].

2.3. Thalidomide (THD). THD is an immunomodulatory drug, used mainly in the treatment of specific tumors. It is usually considered the last-line therapy for GIBD, albeit its use is well-documented [52]. The immunomodulatory effects of THD are due to the reduction in levels of TNF-[alpha] because of degradation of its encoding mRNA [53]. A pilot study [54] and three open studies [55-57] have demonstrated THD effectiveness in the treatment of BD with mucocutaneous involvement as well as in CD enteric involvement [58].

Yasui et al. reported the benefits of THD in 7 juvenile-onset patients with severe, recurrent GIBD who had previously failed immunosuppressant treatments and developed significant CC toxicity. THD was given at the initial dose of 2 mg/kg per day and was increased to 3 mg/kg per day if necessary. All patients showed dramatic improvement in clinical symptoms, and CC were successfully withdrawn [59]. Yet, the efficacy of THD has been also reported on four BD patients with relapsing gastrointestinal disease who required the frequent use of systemic CC. Three out of the four patients had a clinical improvement on THD treatment and all discontinued CC therapy suggesting that THD could be considered a therapeutic option for treatment of refractory GIBD [60]. More recently Hatemi et al. described their experience on 13 patients with GIBD refractory to the conventional therapy who were treated with TNF-[alpha] antagonists and/or THD; a clinical and endoscopic remission was obtained in 10 out of 13 patients (about 75% of the cases) proving the favourable response with anti-TNF-[alpha] agents and/or THD in GIBD [61].

Despite the efficacy and safety profile reported in several bodies of evidence of the literature [59, 62, 63], more data from clinical trials are necessary to define the proper use of this widely known teratogenic drug.

2.4. Other Immunomodulators. Thiopurines including 6-mercaptopurine (6-MP) and its prodrug AZA have been traditionally thought to decrease reoperation rate in patients with GIBD who already had undergone surgical interventions [25]. The initial dose of AZA is 25-50 mg/day with gradual increase every 2-4 week to 2.0-2.5mg/kg [64]. Similarly the starting dose of 6-MP is 0.5 mg/kg escalated every 2-4 weeks to an optimal dosing regimen of 1.0-1.5 mg/kg. Data on the questionable effectiveness of thiopurine treatment in patients with GIBD derive from a retrospective analysis aimed at investigating predictors of clinical relapse in 67 patients with GIBD receiving thiopurine maintenance therapy; a cumulative relapse rates of 5.8%, 28.7%, 43.7%, and 51.7% at 1, 2, 3, and 5 years, respectively, after remission were recorded. Although thiopurine therapy has proven to be relatively effective for maintenance of remission in GIBD, a younger age at diagnosis (<25 years) and a lower hemoglobin level (<11g/dL) were associated with a poor response to this treatment [65].

Yet, a recent retrospective observational study was carried out to assess the efficacy of postoperative thiopurine therapy in 77 patients with GIBD; although a lower postoperative recurrence was found in patients who received thiopurines than those taking 5-ASA, the rates of reoperation, readmission, and death were not significantly different between the 5-ASA and thiopurine groups [66].

Evidence concerning the efficacy of methotrexate combined with infliximab (IFX) in refractory entero-BD has been also reported in 9 out of 10 patients who experienced the disappearance of ileocecal ulcerations at 12 months of therapy [67]. In addition, anecdotal case reports describing the effectiveness of tacrolimus [68] and chlorambucil [69] for treating intestinal lesions in BD have been also described.

Interferon (IFN) is a cytokine able to render cells resistant to infection by many viruses. It was introduced for the treatment of BD by Tsambaos in 1986, because of its antiviral and antiproliferative properties [70]. The most impressive results have been achieved for severe and/or refractory ocular manifestations; however IFN-a could represent a promising treatment option for neurologic and gastrointestinal involvement in BD [71]. In this regard Grimbacher et al. reported a complete remission of BD retinal infiltrates and BD-related colitis after treatment with IFN-a [72], and similar proofs supporting the benefits of IFN-a also derive from the study by Monastirli et al. [73].

During the last few decades intravenous immunoglobulins (IVIg) have been increasingly administered for a wide number of autoimmune and systemic inflammatory diseases. To the best of our knowledge, IVIg have so far been evaluated in few patients with GIBD. In this regard, the efficacy of IVIg has been reported in a patient with BD-related colitis who initially had failed under CC and immunosuppressive therapy [19], as well as in a patient with GIBD complicated by the presence of immune deficiency [74]. More recently, four BD patients, one of whom suffered from neurological and gastrointestinal involvement, refractory to standard treatments and responsive to IVIg therapy have also been described [75].

2.5. Biological Drugs. Several data suggest a role of TNF[alpha] in the pathogenesis of BD; a remarkable upregulation of TNF-[alpha] and soluble TNF receptors [76] as well as a great amount of [gamma][delta]+ T cells producing TNF were found in the peripheral blood of patients with active disease [77]. Currently, the monoclonal antibodies anti-TNF-[alpha] IFX [78] and adalimumab (ADA) [79] along with the human TNF receptor p75 Fc fusion protein etanercept (ETN) [80] have been advocated for the treatment of different BD manifestations. The administration schedule of IFX for treating GIBD is adopted from the regimen employed in the management of CD (5 mg/kg intravenous at weeks 0,2, and 6) [81]. A clinical remission of intestinal BD lesions and the rapid healing of ulcers after treatment with IFX have been described in several case reports (10, 40, 82-90). The short and long-term effects of IFX on the clinical course and intestinal manifestations of BD were assessed by abdominal computed tomography and colonoscopy in ten patients with entero-BD refractory to the conventional therapies; all patients showed improvement of gastrointestinal symptoms and disease-associated complications within 4 weeks. Furthermore, the rate of disappearance of ileocecal ulcerations was 50% (5/10 patients) at 6 months and 90% (9/10 patients) at 12 months [67]. On the contrary, the results derived from a retrospective observational study enrolling 43 patients with GIBD were not entirely encouraging; in this context, IFX chosen as optional treatment in 7 patients refractory to conventional therapies led to clinical remission only in one case (14%) [82].

Yet, a Korean multicenter retrospective study aimed at investigating the response to IFX in 28 patients with GIBD showed a clinical response rate of 64.3% with a clinical remission rate of 28.6% at week 4, following IFX infusion. Furthermore, an older age at diagnosis ([greater than or equal to]40 years), the female sex, a longer disease duration ([greater than or equal to]5 years) as well as the concomitant immunomodulator use, and achievement of remission at week 4 were regarded as predictive factors of sustained response [83].

The efficacy of IFX in GIBD has also been corroborated by a retrospective cohort study on 15 patients with active disease refractory to conventional medications. 80% of patients exhibited a good response to IFX and 53% of them achieved remission after 10 weeks. Moreover 64% and 50% of patients maintained the response to IFX at 12 and 24 months, respectively [84]. More recently an open-label single-arm phase 3 study carried out on 18 BD patients including 11 with GIBD suggested that IFX was able to induce a clinical amelioration along with decrease in C-reactive protein levels after week 2. Consistently, the healing of the main intestinal ulcers was found in more than 80% of these patients after week 14. Interestingly, 3 patients who had loss of response to IFX showed complete resolution of symptoms by increasing its dosage to 10 mg/kg [85].

Finally, an interventional open-label single-arm study testing the efficacy of IFX by assessing the mean decrease in DAIBD score in patients with active intestinal disease refractory to conventional therapies is currently recruiting participants (ClinicalTrials.gov, NCT02505568).

To date, few data are available regarding ADA efficacy in GIBD, although the proofs of its usefulness are increasing [86-88]. In this regard, ADA has been already successfully used in three patients with BD-related colitis/esophageal ulcers [89] and for the first time in the context of a familial case of GIBD [90].

However, the most consistent evidence regarding ADA efficacy derives from a phase 3, multicenter, open-label uncontrolled study evaluating Japanese patients with active intestinal BD nonresponsive to standard therapies. Twenty patients received induction treatment with ADA (160 mg at week 0, baseline, and 80 mg at week 2, followed by 40 mg every other week for 52 weeks); a composite index, combining GI symptoms and endoscopic assessments, was used to evaluate the efficacy of treatment. A marked improvement, defined as values [less than or equal to]1 for both the global GI symptoms and endoscopic assessment scores, was seen in 60% of patients at week 52. Interestingly 20% of patients achieved a complete remission, defined as global GI symptoms and endoscopic scores of 0, at weeks 24 and 52, suggesting that ADA was an effective therapy to induce and maintain clinical improvement and remission in patients with GIBD [91]. More recently, a retrospective study on 8 BD patients with intestinal BD, confirmed the long-term (52 weeks) efficacy and safety of ADA for the treatment of GIBD [92]. Of note, two prospective observational studies (clinicaltrials.gov NCT02687828 and NCT01960790) testing the safety and efficacy of ADA for the treatment of GIBD are now ongoing.

Less experience has been gained focusing on the management of GIBD with ETA treatment [93]. Recently Ma et al. have proven the superiority of ETA in GIBD as compared to conventional therapy, assessing the disappearance of intestinal ulcers confirmed by endoscopy. The healing rate of intestinal ulcers in the group treated with ETA (19 patients) was 89.47%, whereas in the group undergoing conventional treatments (35 patients) it was 51.42%. Therefore, these results proved a better curative effect of ETA as compared to conventional therapies [94].

Although the employment of anti-IL-1 agents on various BD manifestations has been well-documented [95-97], limited data are available for their efficacy in GIBD, being represented only by single-case reports and small case series that describe a clinical amelioration of symptoms without a clear improvement of organic lesions [15-17]. Similarly, albeit IL-6 could be a relevant therapeutic target for refractory BD and its activity can be blocked using the anti-IL-6 receptor antibody tocilizumab [98, 99], inconsistent are the literature data concerning the efficacy of this biologic drug in managing BD clinical manifestations that differ from the neurological ones [100-102].

2.6. Stem Cell Transplantation. Haematopoietic stem cell transplantation (HSCT) has been used in the treatment of severe autoimmune and inflammatory conditions unresponsive to conventional therapy. Although several treatment options, including biologic agents, are till now available for BD management, there is still an unmet need for more effective therapies for patients who are refractory to conventional treatments. In this regard encouraging results derive from several case reports describing HSCT in GIBD patients transplanted for accompanying haematological conditions. A complete remission of GI findings was observed after HSCT and there was no need to treat patients with any medications [103, 104]. This evidence suggests that HSCT may be an effective alternative in BD patients with severe organ involvement, especially GI involvement refractory to immunosuppressives. However, one must make sure that the benefit outweighs the risks when developing a management strategy for these patients. Since HSCT can be a life-threatening procedure, mostly autologous transplants should be preferred to those allogenic which may lead to major complications such as infections, GVHD, and hepatic, renal, and pulmonary damage [105].

2.7. Surgery. Surgery is indicated when patients with GIBD are refractory to medical treatment or serious complications, such as when gastrointestinal bleeding, perforation, fistulae, obstructions, and abdominal masses occur [106]. Bowel perforation is one of the most feared complications of GIBD. In this regard a retrospective study analysing free bowel wall perforation in 129 subjects with GIBD showed that 25.6% of patients experienced surgery for bowel perforation. Of them, 42.4% showed postoperative recurrence and 33.3% underwent reoperation. In addition, a younger age at diagnosis ([less than or equal to]25 years), an experience of prior laparotomy, and the presence of volcano-shaped ulcers were regarded as independent risk factors for free bowel perforation [107]. Resection involving sufficient margin and including normal bowel has been widely accepted in surgery [108,109]. However, some bodies of evidence have proven that resection length is not related to postoperative recurrence in patients with GIBD, prospecting a less invasive surgical approach [39,106]. Intestinal leakage, perforation, and fistula formation seems to occur more frequently at the anastomotic site; consequently, the creation of a stoma is preferred over primary anastomosis [18].

3. Conclusions

BD is a complex syndrome characterized by significant heterogeneity of clinical manifestations with usually frequent relapses. GIBD is one of the most severe manifestations of this disease, causing serious complications such as perforation and gastrointestinal bleeding. GIBD shares many clinical features with inflammatory bowel diseases, and for this reason it represents a pitfall for physicians regarding differential diagnosis at the first presentation. Nevertheless, a careful evaluation of endoscopic findings may help in the diagnostic interpretation, whereas the endoscopic biopsy is necessary to confirm a histopathologic diagnosis. Treatments have been largely unsatisfactory, creating significant unmet needs, and the lack of evidence-based treatment makes the management of GIBD very challenging. Currently, several immunosuppressive drugs such as SSZ, CC, and AZA are generally well-tolerated but often associated with increased risk of disease relapses as a result of which surgery is required in many patients. In the last years, new pathogenetic hypotheses supported the use of biotechnological drugs, mostly TNF-[alpha] inhibitors, which represent new tools in the therapeutic armamentarium for managing patients with GIBD, particularly those who are resistant to conventional immunosuppressant drugs. On this basis the main goal of treatment should be aimed at avoiding and preventing the feared complications of GIBD that endanger the life of these patients.

http://dx.doi.org/10.1155/2017/1460491

Competing Interests

The authors declare that there is no conflict of interests regarding the publication of this paper.

Authors' Contributions

Giuseppe Lopalco and Donato Rigante equally contributed to this manuscript.

References

[1] D. G. James, "Behcet's syndrome," New England Journal of Medicine, vol. 301, no. 8, pp. 431-432, 1979.

[2] V. G. Kaklamani, G. Vaiopoulos, and P. G. Kaklamanis, "Behcet's disease," Seminars in Arthritis and Rheumatism, vol. 27, no. 4, pp. 197-217, 1998.

[3] T. Sakane, M. Takeno, N. Suzuki, and G. Inaba, "Behcet's disease," New England Journal of Medicine, vol. 341, no. 17, pp. 1284-1291, 1999.

[4] D. H. Verity, J. E. Marr, S. Ohno, G. R. Wallace, and M. R. Stanford, "Behcet's disease, the Silk Road and HLA-B51: historical and geographical perspectives," Tissue Antigens, vol. 54, no. 3, pp. 213-220, 1999.

[5] L. Cantarini, G. Lopalco, F. Caso et al., "Effectiveness and tuberculosis-related safety profile of interleukin-1 blocking agents in the management of Behcet's disease," Autoimmunity Reviews, vol. 14, no. 1, pp. 1-9, 2015.

[6] G. Lopalco, O. M. Lucherini, A. Vitale et al., "Putative role of serum amyloid-A and proinflammatory cytokines as biomarkers for Behcet's disease," Medicine (Baltimore), vol. 94, no. 42, p. e1858, 2015.

[7] F. Caso, L. Costa, D. Rigante et al., "Biological treatments in behcet's disease: beyond anti-TNF therapy," Mediators of Inflammation, vol. 2014, Article ID 107421,14 pages, 2014.

[8] M. Pineton de Chambrun, B. Wechsler, G. Geri, P. Cacoub, and D. Saadoun, "New insights into the pathogenesis of Behcet's disease," Autoimmunity Reviews, vol. 11, no. 10, pp. 687-698, 2012.

[9] Z. Y. Zhou, S. L. Chen, N. Shen, and Y. Lu, "Cytokines and Behcet's disease," Autoimmunity Reviews, vol. 11, no. 10, pp. 699-704, 2012.

[10] S. P L. Travis, M. Czajkowski, D. P B. McGovern, R. G. P Watson, and A. L. Bell, "Treatment of intestinal Behcet's syndrome with chimeric tumour necrosis factor a antibody," Gut, vol. 49, no. 5, pp. 725-728, 2001.

[11] P. V. Hassard, S. W. Binder, V. Nelson, and E. A. Vasiliauskas, "Anti-tumor necrosis factor monoclonal antibody therapy for gastrointestinal behcet's disease: a case report," Gastroenterology, vol. 120, no. 4, pp. 995-999, 2001.

[12] M. T. Kram, L. D. May, S. Goodman, and S. Molinas, "Behcet's ileocolitis: successful treatment with tumor necrosis factor-alpha antibody (infliximab) therapy. Report of a case," Diseases of the Colon and Rectum, vol. 46, no. 1, pp. 118-121, 2003.

[13] S. Hirohata, H. Kikuchi, T. Sawada et al., "Clinical characteristics of neuro-Behcet's disease in Japan: a multicenter retrospective analysis," Modern Rheumatology, vol. 22, no. 3, pp. 405-413, 2012.

[14] E. H. Kim, M.-J. Park, S. Park, and E.-S. Lee, "Increased expression of the NLRP3 inflammasome components in patients with Behcet's disease," Journal of Inflammation (United Kingdom), vol. 12, no. 1, article 41, 2015.

[15] L. Cantarini, A. Vitale, P. Scalini et al., "Anakinra treatment in drug-resistant Behcet's disease: a case series," Clinical Rheumatology, vol. 34, no. 7, pp. 1293-1301, 2015.

[16] A. Vitale, D. Rigante, F. Caso et al., "Inhibition of interleukin-1 by canakinumab as a successful mono-drug strategy for the treatment of refractory behcet's disease: a case series," Dermatology, vol. 228, no. 3, pp. 211-214, 2014.

[17] L. Cantarini, A. Vitale, M. Borri, M. Galeazzi, and R. Franceschini, "Successful use of canakinumab in a patient with resistant Behcet's disease," Clinical and Experimental Rheumatology, vol. 30, supplement 72, p. S115, 2012.

[18] Y. Bayraktar, E. Ozaslan, and D. H. Van Thiel, "Gastrointestinal manifestations of Behcet's disease," Journal of Clinical Gastroenterology, vol. 30, no. 2, pp. 144-154, 2000.

[19] I. L. P. Beales, "Gastrointestinal involvement in Behcet's syndrome," American Journal of Gastroenterology, vol. 93, no. 12, 2633 pages, 1998.

[20] S. E. Marshall, "Behcet's disease," Best Practice & Research: Clinical Rheumatology, vol. 18, no. 3, pp. 291-311, 2004.

[21] D. H. Kim and J. H. Cheon, "Intestinal behcet's disease: a true inflammatory bowel disease or merely an intestinal complication of systemic vasculitis?" Yonsei Medical Journal, vol. 57, no. 1, pp. 22-32, 2016.

[22] D. Mendes, M. Correia, M. Barbedo et al., "Behcet's disease-- a contemporary review," Journal of Autoimmunity, vol. 32, no. 3-4, pp. 178-188, 2009.

[23] P. P. Sfikakis, N. Markomichelakis, E. Alpsoy et al., "Anti-TNF therapy in the management of Behcet's disease--review and basis for recommendations," Rheumatology, vol. 46, no. 5, pp. 736-741, 2007.

[24] B. E. Sands, "From symptom to diagnosis: clinical distinctions among various forms of intestinal inflammation," Gastroenterology, vol. 126, no. 6, pp. 1518-1532, 2004.

[25] I. J. Choi, J. S. Kim, S. D. Cha et al., "Long-term clinical course and prognostic factors in intestinal Behcet's disease," Diseases of the Colon and Rectum, vol. 43, no. 5, pp. 692-700, 2000.

[26] C. R. Lee, W. H. Kim, Y. S. Cho et al., "Colonoscopic findings in intestinal Behcet's disease," Inflammatory Bowel Diseases, vol. 7, no. 3, pp. 243-249, 2001.

[27] J. H. Cheon and W. H. Kim, "An update on the diagnosis, treatment, and prognosis of intestinal Behcet's disease," Current Opinion in Rheumatology, vol. 27, no. 1, pp. 24-31, 2015.

[28] S. Yurdakul, N. Timiner, I. Yurdakul, V. Hamuryudan, and H. Yazici, "Gastrointestinal involvement in Behcet's syndrome: a controlled study," Annals of the Rheumatic Diseases, vol. 55, no. 3, pp. 208-210, 1996.

[29] S. Mori, A. Yoshihira, H. Kawamura, A. Takeuchi, T. Hashimoto, and G. Inaba, "Esophageal involvement in Behcet's disease," The American Journal of Gastroenterology, vol. 78, no. 9, pp. 548-553, 1983.

[30] E. L. Grigg, S. Kane, and S. Katz, "Mimicry and deception in inflammatory bowel disease and intestinal Behcet disease," Gastroenterology and Hepatology, vol. 8, no. 2, pp. 103-112, 2012.

[31] L. Ning-Sheng, L. Ruay-Sheng, and T. Kuo-Chih, "High frequency of unusual gastric/duodenal ulcers in patients with Behcet's disease in Taiwan: a possible correlation of MHC molecules with the development of gastric/duodenal ulcers," Clinical Rheumatology, vol. 24, no. 5, pp. 516-520, 2005.

[32] E. C. Ebert, "Gastrointestinal manifestations of Behcet's disease," Digestive Diseases and Sciences, vol. 54, no. 2, pp. 201-207, 2009.

[33] A. B. Chin and A. S. Kumar, "Behcet colitis," Clinics in Colon and Rectal Surgery, vol. 28, no. 2, pp. 99-102, 2015.

[34] J. S. Kim, S. H. Lim, I. J. Choi et al., "Prediction of the clinical course of Behcet's colitis according to macroscopic classification by colonoscopy," Endoscopy, vol. 32, no. 8, pp. 635-640, 2000.

[35] M. J. Chung, J. H. Cheon, S. U. Kim et al., "Response rates to medical treatments and long-term clinical outcomes of nonsurgical patients with intestinal Behcet disease," Journal of Clinical Gastroenterology, vol. 44, no. 6, pp. e116-e122, 2010.

[36] S. K. Lee, B. K. Kim, T. I. Kim, and W. H. Kim, "Differential diagnosis of intestinal Beh9cet's disease and Crohn's disease by colonoscopic findings," Endoscopy, vol. 41, no. 1, pp. 9-16, 2009.

[37] J. H. Cheon, D. S. Han, J. Y. Park et al., "Development, validation, and responsiveness of a novel disease activity index for intestinal Behcet's disease," Inflammatory Bowel Diseases, vol. 17, no. 2, pp. 605-613, 2011.

[38] G. Hatemi, A. Silman, D. Bang et al., "Management of Behcet disease: a systematic literature review for the European League Against Rheumatism evidence-based recommendations for the management of Behcet disease," Annals of the Rheumatic Diseases, vol. 68, no. 10, pp. 1528-1534, 2009.

[39] M. Iida, H. Kobayashi, T. Matsumoto et al., "Postoperative recurrence in patients with intestinal Behcet's disease," Diseases of the Colon & Rectum, vol. 37, no. 1, pp. 16-21, 1994.

[40] J. H. Lee, T. N. Kim, S. T. Choi et al., "Remission of intestinal Behcet's disease treated with anti-tumor necrosis factor a monoclonal antibody (Infliximab)," Korean Journal of Internal Medicine, vol. 22, no. 1, pp. 24-27, 2007

[41] R. Brik, H. Shamali, and R. Bergman, "Successful thalidomide treatment of severe infantile Behcet disease," Pediatric Dermatology, vol. 18, no. 2, pp. 143-145, 2001.

[42] J. Fallingborg and J. Laustsen, "Colitis of Behcet's syndrome," Acta Medica Scandinavica, vol. 215, no. 4, pp. 397-399, 1984.

[43] K. Toda, Y. Shiratori, M. Yusuda et al., "Therapeutic effect of intraarterial prednisolone injection in severe intestinal Behcet's disease," Journal of Gastroenterology, vol. 37, no. 10, pp. 844-848, 2002.

[44] J. J. Park, W. H. Kim, and J. H. Cheon, "Outcome predictors for intestinal Behcet's disease," Yonsei Medical Journal, vol. 54, no. 5, pp. 1084-1090, 2013.

[45] S. Narum, T. Westergren, and M. Klemp, "Corticosteroids and risk of gastrointestinal bleeding: a systematic review and meta-analysis," BMJ Open, vol. 4, no. 5, Article ID e004587, 2014.

[46] K. Kobayashi, F. Ueno, S. Bito et al., "Development of consensus statements for the diagnosis and management of intestinal Behcet's disease using a modified Delphi approach," Journal of Gastroenterology, vol. 42, no. 9, pp. 737-745, 2007

[47] M. H. Houman and K. Hamzaoui, "Promising new therapies for Behcet's disease," European Journal of Internal Medicine, vol. 17, no. 3, pp. 163-169, 2006.

[48] T. Sonta, Y. Araki, M. Kubokawa et al., "The beneficial effect of mesalazine on esophageal ulcers in intestinal Behcet's disease," Journal of Clinical Gastroenterology, vol. 30, no. 2, pp. 195-197, 2000.

[49] M. Matsukawa, T. Yamasaki, T. Kouda, and M. Kurihara, "Endoscopic therapy with absolute ethanol for postoperative recurrent ulcers in intestinal Behcet's disease, and simple ulcers," Journal of Gastroenterology, vol. 36, no. 4, pp. 255-258, 2001.

[50] Y. S. Jung, S. P. Hong, T. I. Kim, W. H. Kim, and J. H. Cheon, "Long-term clinical outcomes and factors predictive of relapse after 5-aminosalicylate or sulfasalazine therapy in patients with intestinal Behcet disease," Journal of Clinical Gastroenterology, vol. 46, no. 5, pp. e38-e45, 2012.

[51] I. Hatemi, S. N. Esatoglu, G. Hatemi, Y. Erzin, H. Yazici, and A. F. Celik, "Characteristics, treatment, and long-term outcome of gastrointestinal involvement in behcet's syndrome: a strobe-compliant observational study from a dedicated multidisciplinary center," Medicine (United States), vol. 95, no. 16, Article ID e3348, 2016.

[52] M. Sayarlioglu, M. C. Kotan, N. Topcu, I. Bayram, H. Arslanturk, and A. Gul, "Treatment of recurrent perforating intestinal ulcers with thalidomide in Behccet's disease," Annals of Pharmacotherapy, vol. 38, no. 5, pp. 808-811, 2004.

[53] A. L. Moreira, E. P. Sampaio, A. Zmuidzinas, P. Frindt, K. A. Smith, and G. Kaplan, "Thalidomide exerts its inhibitory action on tumor necrosis factor a by enhancing mRNA degradation," Journal of Experimental Medicine, vol. 177, no. 6, pp. 1675-1680, 1993.

[54] B. De Wazieres, H. Gil, N. Magy, S. Berthier, D. A. Vuitton, and J. L. Dupond, "Treatment of recurrent oro-genital ulceration with low dose of thalidomide. Pilot study in 17 patients," Revue de Medecine Interne, vol. 20, no. 7, pp. 567-570, 1999.

[55] J. M. M. Gardner-Medwin, N. J. Smith, and R. J. Powell, "Clinical experience with thalidomide in the management of severe oral and genital ulceration in conditions such as Behccet's disease: use of neurophysiological studies to detect thalidomide neuropathy," Annals of the Rheumatic Diseases, vol. 53, no. 12, pp. 828-832, 1994.

[56] T. Saylan and I. Saltik, "Thalidomide in the treatment of Behcet's syndrome," Archives of Dermatology, vol. 118, no. 8, 1982.

[57] M. H. Hamza, "Treatment of Behcet's disease with thalidomide," Clinical Rheumatology, vol. 5, no. 3, pp. 365-371, 1986.

[58] C. Bariol, A. P. Meagher, C. R. Vickers et al., "Thalidomide for inflammatory bowel disease: early studies on the safety and efficacy of thalidomide for symptomatic inflammatory bowel disease," Journal of Gastroenterology and Hepatology, vol. 17, no. 2, pp. 135-139, 2002.

[59] K. Yasui, N. Uchida, Y. Akazawa et al., "Thalidomide for treatment of intestinal involvement of juvenile-onset Behcet disease," Inflammatory Bowel Diseases, vol. 14, no. 3, pp. 396-400, 2008.

[60] H. J. Lee, J. H. Cheon, K. J. Lee et al., "Clinical experience of thalidomide in the treatment of korean patients with intestinal Behccet's disease: pilot experience in a single center," Intestinal Research, vol. 8, no. 1, pp. 63-69, 2010.

[61] I. Hatemi, G. Hatemi, O. N. Pamuk, Y. Erzin, and A. F. Celik, "TNF-alpha antagonists and thalidomide for the management of gastrointestinal Behccet's syndrome refractory to the conventional treatment modalities: a case series and review of the literature," Clinical and Experimental Rheumatology, vol. 33, no. 6, supplement 94, pp. S129-S137, 2015.

[62] O. Patey, N. Charasz, A. M. Roucayrol, J. E. Malkin, and C. Lafaix, "Thalidomide and ulcerative colitis in Behccet's disease," Gastroenterologie Clinique et Biologique, vol. 13, no. 1, article no. 104, 1989.

[63] H. Larsson, "Treatment of severe colitis in Behcet's syndrome with thalidomide (CG-217)," Journal of Internal Medicine, vol. 228, no. 4, pp. 405-407, 1990.

[64] T. Hisamatsu, F. Ueno, T. Matsumoto et al., "The 2nd edition of consensus statements for the diagnosis and management of intestinal Behcet's disease: indication of anti-TNF[alpha] monoclonal antibodies," Journal of Gastroenterology, vol. 49, no. 1, pp. 156-162, 2014.

[65] Y. S. Jung, J. H. Cheon, S. P. Hong, T. I. Kim, and W. H. Kim, "Clinical outcomes and prognostic factors for thiopurine maintenance therapy in patients with intestinal Behccet's disease," Inflammatory Bowel Diseases, vol. 18, no. 4, pp. 750-757, 2012.

[66] H. W. Lee, J. H. Cheon, H. J. Lee et al., "Postoperative effects of thiopurines in patients with intestinal Behccet's disease," Digestive Diseases and Sciences, vol. 60, no. 12, pp. 3721-3727, 2015.

[67] S. Iwata, K. Saito, K. Yamaoka et al., "Efficacy of combination therapy of anti-TNF-[alpha] antibody infliximab and methotrexate in refractory entero-Behcet's disease," Modern Rheumatology, vol. 21, no. 2, pp. 184-191, 2011.

[68] K. Matsumura, H. Nakase, and T. Chiba, "Efficacy of oral tacrolimus on intestinal Behcet's disease," Inflammatory Bowel Diseases, vol. 16, no. 2, pp. 188-189, 2010.

[69] R. C. Wong, C. N. Ellis, and L. A. Diaz, "Behcet's disease," International Journal of Dermatology, vol. 23, no. 1, pp. 25-32, 1984.

[70] D. Tsambaos, D. Eichelberg, and M. Goos, "Behccet's syndrome: treatment with recombinant leukocyte alpha-interferon," Archives of Dermatological Research, vol. 278, no. 4, pp. 335-336, 1986.

[71] I. Kotter, I. Gunaydin, M. Zierhut, and N. Stubiger, "The use of interferon a in Behcet disease: review of the literature," Seminars in Arthritis and Rheumatism, vol. 33, no. 5, pp. 320-335, 2004.

[72] B. Grimbacher, B. Wenger, P. Deibert, T. Ness, I. Koetter, and H. H. Peter, "Loss of vision and diarrhoea," Lancet, vol. 350, no. 9094, p. 1818, 1997.

[73] A. Monastirli, E. Chroni, S. Georgiou et al., "Interferon-a treatment for acute myelitis and intestinal involvement in severe Behcet's disease," QJM, vol. 103, no. 10, pp. 787-790, 2010.

[74] B. Shutty, K. J. Garg, D. Swender, L. Chernin, H. Tcheurekdjian, and R. Hostoffer, "Optimal use of ivig in a patient with Behcet syndrome and common variable immunodeficiency," Annals of Allergy, Asthma and Immunology, vol. 109, no. 1, article no. 84, 2012.

[75] L. Cantarini, M. L. Stromillo, A. Vitale et al., "Efficacy and safety of intravenous immunoglobulin treatment in refractory Behcet's disease with different organ involvement: a case series," Israel Medical Association Journal, vol. 18, no. 3-4, pp. 238-242, 2016.

[76] G. Triolo, A. Accardo-Palumbo, F. Dieli et al., "V[gamma]9/V[delta]2 T lymphocytes in Italian patients with Behcet's disease--evidence for expansion, and tumour necrosis factor receptor II and interleukin-12 receptor [[beta].sub.1] expression in active," Arthritis Research and Therapy, vol. 5, no. 5, 2003.

[77] J. Freysdottir, L. Hussain, I. Farmer, S.-H. Lau, and F. Fortune, "Diversity of [gamma][delta] T cells in patients with Behcet's disease is indicative of polyclonal activation," Oral Diseases, vol. 12, no. 3, pp. 271-277, 2006.

[78] N. Pipitone, I. Olivieri, A. Padula et al., "Infliximab for the treatment of neuro-Behcet's disease: a case series and review of the literature," Arthritis Care and Research, vol. 59, no. 2, pp. 285-290, 2008.

[79] D. Perra, M. A. Alba, J. L. Callejas et al., "Adalimumab for the treatment of Behcet's disease: experience in 19 patients," Rheumatology, vol. 51, no. 10, Article ID kes130, pp. 1825-1831, 2012.

[80] L. Cantarini, I. Tinazzi, P Caramaschi, F. Bellisai, A. Brogna, and M. Galeazzi, "Safety and efficacy of etanercept in children with juvenile-onset Behcet's disease," International Journal of Immunopathology and Pharmacology, vol. 22, no. 2, pp. 551-555, 2009.

[81] R. Caviglia, I. Boskoski, and M. Cicala, "Maintenance treatment with infliximab for the management of Crohn's disease in adults," Biologics, vol. 3, pp. 39-49, 2009.

[82] H. Ideguchi, A. Suda, M. Takeno et al., "Gastrointestinal manifestations of Behcet's disease in Japan: a study of 43 patients," Rheumatology International, vol. 34, no. 6, pp. 851-856, 2014.

[83] J. H. Lee, J. H. Cheon, S. W. Jeon et al., "Efficacy of infliximab in intestinal Behcet's disease: a Korean multicenter retrospective study," Inflammatory Bowel Diseases, vol. 19, no. 9, pp. 1833-1838, 2013.

[84] H. Kinoshita, R. Kunisaki, H. Yamamoto et al., "Efficacy of infliximab in patients with intestinal Behccet's disease refractory to conventional medication," Internal Medicine, vol. 52, no. 17, pp. 1855-1862, 2013.

[85] T. Hibi, S. Hirohata, H. Kikuchi et al., "Infliximab therapy for intestinal, neurological, and vascular involvement in Behcet disease: efficacy, safety, and pharmacokinetics in a multicenter, prospective, open-label, single-arm phase 3 study," Medicine (United States), vol. 95, no. 24, p. e3863, 2016.

[86] M. Kimura, Y. Tsuji, M. Iwai et al., "Usefulness of adalimumab for treating a case of intestinal Behcet's disease with trisomy 8 myelodysplastic syndrome," Intestinal Research, vol. 13, no. 2, pp. 166-169, 2015.

[87] T. Mizoshita, S. Tanida, and T. Joh, "Adalimumab treatment in intestinal Behccet's disease: relationship with ectopic mucin 5AC glycoprotein expression and endoscopic improvement," Digestive and Liver Disease, vol. 47, no. 11, pp. 991-992, 2015.

[88] M. Della Maggiora, A. Banos, V. Paolini, D. Florio, L. Takashima, and P. Aicardi, "Behccet's disease with intestinal perforation: a case report," Reumatologia Clinica, 2016.

[89] J. A. M. Van Laar, T. Missotten, P L. A. Van Daele, A. Jamnitski, G. S. Baarsma, and P. M. Van Hagen, "Adalimumab: a new modality for Behcet's disease?" Annals of the Rheumatic Diseases, vol. 66, no. 4, pp. 565-566, 2007.

[90] C. De Cassan, B. De Vroey, C. Dussault, E. Hachulla, S. Buche, and J.-F. Colombel, "Successful treatment with adalimumab in a familial case of gastrointestinal Behcet's disease," Journal of Crohn's and Colitis, vol. 5, no. 4, pp. 364-368, 2011.

[91] S. Tanida, N. Inoue, K. Kobayashi et al., "Adalimumab for the treatment of japanese patients with intestinal Behcet's disease," Clinical Gastroenterology and Hepatology, vol. 13, no. 5, pp. 940.e3-948.e3, 2015.

[92] S. Tanida, T. Mizoshita, H. Nishie et al., "Long-Term efficacy of adalimumab in patients with intestinal Behccet's disease: eight consecutive cases," Journal of Clinical Medicine Research, vol. 8, no. 4, pp. 334-337, 2016.

[93] S. Watanabe, T. Aizawa-Yashiro, K. Tsuruga, M. Kinjo, E. Ito, and H. Tanaka, "A young girl with refractory intestinal Behccet's disease: a case report and review of literatures on pediatric cases who received an anti-tumor necrosis factor agent," Rheumatology International, vol. 33, no. 12, pp. 3105-3108, 2013.

[94] D. Ma, C.-J. Zhang, R.-P Wang, L. Wang, and H. Yang, "Etanercept in the treatment of intestinal Behcet's disease," Cell Biochemistry and Biophysics, vol. 69, no. 3, pp. 735-739, 2014.

[95] Y. Bilginer, N. A. Ayaz, and S. Ozen, "Anti-IL-1 treatment for secondary amyloidosis in an adolescent with FMF and Behcet's disease," Clinical Rheumatology, vol. 29, no. 2, pp. 209-210, 2010.

[96] C. Botsios, P. Sfriso, A. Furlan, L. Punzi, and C. A. Dinarello, "Resistant Behccet disease responsive to anakinra," Annals of Internal Medicine, vol. 149, no. 4, pp. 284-286, 2008.

[97] G. Emmi, E. Silvestri, A. M. Cameli et al., "Anakinra for resistant Behccet uveitis: why not?" Clinical and Experimental Rheumatology, vol. 31, no. 77, pp. S152-S153, 2013.

[98] K. Hamzaoui, A. Hamzaoui, F. Guemira, M. Bessioud, M. Hamza, and K. Ayed, "Cytokine profile in Behcet's disease patients: relationship with disease activity," Scandinavian Journal of Rheumatology, vol. 31, no. 4, pp. 205-210, 2002.

[99] T. Hirano, N. Ohguro, S. Hohki et al., "A case of Behcet's disease treated with a humanized anti-interleukin-6 receptor antibody, tocilizumab," Modern Rheumatology, vol. 22, no. 2, pp. 298-302, 2012.

[100] L. Cantarini, G. Lopalco, A. Vitale et al., "Paradoxical mucocutaneous flare in a case of Behccet's disease treated with tocilizumab," Clinical Rheumatology, vol. 34, no. 6, pp. 1141-1143, 2015.

[101] G. Emmi, E. Silvestri, D. Squatrito, L. Emmi, L. Cantarini, and D. Prisco, "Tocilizumab-induced exacerbation of mucosal ulcers in a patient with multi-refractory Behccet's disease," Seminars in Arthritis and Rheumatism, vol. 46, no. 1, pp. e1-e2, 2016.

[102] A. Deroux, C. Chiquet, and L. Bouillet, "Tocilizumab in severe and refractory Behcet's disease: four cases and literature review," Seminars in Arthritis and Rheumatism, vol. 45, no. 6, pp. 733-737, 2016.

[103] M.-H. Kook, H.-Y. Yhim, N.-R. Lee et al., "Successful treatment of myelodysplastic syndrome and behcet colitis after allogeneic hematopoietic stem cell transplantation," Korean Journal of Internal Medicine, vol. 29, no. 1, pp. 123-125, 2014.

[104] J. K. Ahn, H.-S. Cha, E.-M. Koh et al., "Behcet's disease associated with bone marrow failure in Korean patients: clinical characteristics and the association of intestinal ulceration and trisomy 8," Rheumatology, vol. 47, no. 8, pp. 1228-1230, 2008.

[105] T. Soysal, A. Salihoglu, S. N. Esatoglu et al., "Bone marrow transplantation for Behcet's disease: a case report and systematic review of the literature," Rheumatology, vol. 53, no. 6, pp. 1136-1141, 2014.

[106] Y. S. Jung, J. Y. Yoon, J. H. Lee et al., "Prognostic factors and long-term clinical outcomes for surgical patients with intestinal Behcet's disease," Inflammatory Bowel Diseases, vol. 17, no. 7, pp. 1594-1602, 2011.

[107] C. M. Moon, J. H. Cheon, J. K. Shin et al., "Prediction of free bowel perforation in patients with intestinal Behcet's disease using clinical and colonoscopic findings," Digestive Diseases and Sciences, vol. 55, no. 10, pp. 2904-2911, 2010.

[108] I. Sayek, O. Aran, B. Uzunalimoglu, and E. Hersek, "Intestinal Behcet's disease: surgical experience in seven cases," Hepato-Gastroenterology, vol. 38, no. 1, pp. 81-83, 1991.

[109] S.-J. Chou, V. T.-K. Chen, H.-C. Jan, M. A. Lou, and Y.-M. Liu, "Intestinal perforations in Behcet's disease," Journal of Gastrointestinal Surgery, vol. 11, no. 4, pp. 508-514, 2007.

Giuseppe Lopalco, (1) Donato Rigante, (2) Vincenzo Venerito, (1) Claudia Fabiani, (3) Rossella Franceschini, (4) Michele Barone, (5) Giovanni Lapadula, (1) Mauro Galeazzi, (6) Bruno Frediani, (6) Florenzo Iannone, (1) and Luca Cantarini (6)

(1) Department of Emergency and Organ Transplantation, Rheumatology Unit, University of Bari, Piazza Giulio Cesarell, 70124 Bari, Italy

(2) Institute of Pediatrics, Universita Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli, Largo A. Gemelli 8, 00168 Rome, Italy

(3) Department of Ophthalmology, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy

(4) Ophthalmology and Neurosurgery Department, University of Siena, Viale Bracci 1, 53100 Siena, Italy

(5) Section of Gastroenterology, Department of Emergency and Organ Transplantation, University of Bari, Piazza Giulio Cesare11, 70124 Bari, Italy

(6) Research Center of Systemic Autoinflammatory Diseases and Behcet's Disease Clinic, Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Viale Bracci 1, 53100 Siena, Italy

Correspondence should be addressed to Luca Cantarini; [email protected]

Received 14 September 2016; Accepted 4 January 2017; Published 22 January 2017

Academic Editor: Ronald Gladue
Table 1: Main clinical, endoscopic, and pathological features of
gastrointestinal involvement in Behcet's disease and most common
localization.

                   Behcet's disease           Crohn's disease

                   Anorexia, vomiting,        Anorexia, vomiting,
Gastrointestinal   dyspepsia, diarrhoea,      dyspepsia, diarrhea,
manifestations     abdominal pain, melena,    gastrointestinal
                   hematochezia, fever        bleeding, abdominal
                                              pain, fever

Pathological       Vasculitis of the          Transmural mucosal
                   small veins and venules    inflammation,
features           with deep ulcerations,     inflammatory cell
                   generally                  infiltrate (lymphocytes,
                   without granulomas or      plasma cells) with focal
                   cobblestoning,             crypt irregularity and
                   ischemic perforation,      independent granulomas
                   thrombosis

                   Round or oval ulcers,      Longitudinal ulcers,
                   punched-out lesions        cobblestone appearance,
Endoscopic         with discrete margins      aphthous ulcers showing
findings           (>1cm), focal              longitudinal array
                   distribution (<5 ulcers)

Localization       Terminal ileum,            Small bowel,
                   ileocecal region, colon    upper-gastrointestinal
                                              tract

                   Ulcerative colitis

                   Rectal bleeding,
Gastrointestinal   diarrhoea, tenesmus,
manifestations     abdominal pain,
                   hematochezia, fever

Pathological       Distortion of crypt
features           architecture,
                   crypt abscesses, lamina
                   propria cellular
                   infiltration (plasma cells,
                   eosinophils,
                   lymphocytes),
                   shortening of the crypts,
                   mucin depletion,
                   lymphoid aggregates,
                   erosions or ulcerations

                   Mucosal erythema, fine
                   granularity, loss of
                   vascular marking,
Endoscopic         erosions, ulcers,
findings           spontaneous bleeding,
                   luminal narrowing with
                   pseudopolyps

Localization       Starts in the rectum and
                   extends proximally in a
                   continuous manner
                   through the entire colon

Table 2: Overview of studies derived from the medical literature
reporting treatment indications of gastrointestinal lesions in
Behcet's disease.

Drugs                     Dose                 Authors      Number of
                                                (year)      patients

5-ASA/SSZ             2.4-4 g/day              Jung et       143/292
                                              al. (2012)

                     2-3 mg/kg/day             Yasui et
THD                                           al. (2008)        7

                                              Lee et al.        4
                                                (2010)

             THD 50/100 mg/day IFX 5 mg/kg    Hatemi et       13/64
              every 8 weeks ADA 160 mg at     al. (2015)
              week 0 and 80 mg at week 2,
THD/IFX/     followed by 40 mg every other
ADA/ETA       week ETA 25 mg twice a week

AZA or       AZA 2/2.5 mg/kg/day 6/MP 0.5/     Jung et       67/272
6-MP                 1.5 mg/kg/day            al. (2012)

AZA          AZA 2/2.5 mg/kg/day or 6/MP 1/   Lee et al.       77
or 6-MP      1.5 mg/kg/day vs 5/ASA 3/4 g/      (2015)
vs                        day
5-ASA

MTX+IFX        MTX /FX 3/5 mg/kg every 8       Iwata et        10
                         weeks                al. (2011)

             6 x [10.sup.6] IU per day for    Grimbacher        1
                        14 days                 et al.
INF-                                            (1997)
[alpha]

                3 x [10.sup.6] IU/day 3
                  times/week increased        Monastirli        1
               to 6 x [10.sup.6] IU/day 3       et al.
                       times/week               (2010)

IVIg          400 mg/kg/day for 5 days per    Cantarini        1/4
                         month                  et al.
                                                (2016)

                           --                  Ideguchi       7/43
                                                et al.
                                                (2014)

IFX              5 mg/kg/every 8 weeks        Lee et al.       28
                                                (2013)

                 5 mg/kg every 8 weeks        Kinoshita       15/43
                                                et al.
                                                (2013)

                 5 mg/kg every 8 weeks         Hibi et        11/18
                                              al. (2016)

ADA          160 mg at week 0 and 80 mg at    Tanida et        20
               week 2, followed by 40 mg      al. (2015)
                    every other week

             160 mg at week 0 and 80 mg at    Tanida et         8
               week 2, followed by 40 mg      al. (2016)
                    every other week

ETA                25 mg twice a week         Ma et al.       19/35
                                                (2014)

                 100 mg/day 2 mg/kg/day
ANA            increased to 2.5 mg/kg/day     Cantarini        3/9
                                                et al.
                                                (2013)

              150 mg every 8 weeks 150 mg     Vitale et        2/3
CANA                every six weeks           al. (2013)

TCZ              8 mg/kg/every 4 weeks        Deroux et        3/4
                                              al. (2015)

Drugs          Authors      Type of study            Outcomes
               (year)

5-ASA/SSZ      Jung et      Retrospective       Positive effect in
             al. (2012)     cohort study       maintaining remission

              Yasui et       Case series      Dramatic improvement in
THD          al. (2008)                          clinical symptoms

             Lee et al.      Case series        3/4 patients had a
               (2010)                        clinical improvement and
                                             all discontinued steroid
              Hatemi et                               therapy
             al. (2015)
                            Observational     Remission obtained with
                                study         TNF/[alpha] antagonists
                                              and/or THD in about 75%
THD/IFX/                                             of cases.
ADA/ETA        Jung et
             al. (2012)
AZA or                      Retrospective    Relative good effect for
6-MP         Lee et al.         study        maintenance of remission
               (2015)
AZA                         Retrospective    The rates of reoperation,
or 6-MP                     observational     readmission, and death
vs                              study         were not significantly
5-ASA         Iwata et                         different between the
             al. (2011)                        5-ASA and thiopurine
                                                      groups
             Grimbacher
MTX+IFX        et al.       Observational    Long-term alleviation of
               (1997)           study         entero-BD and excellent
                                                 tolerability with
                                              combination of IFX and
                                                        MTX
             Monastirli
               et al.                          Complete remission of
               (2010)        Case report         Behqet's retinal
INF-                                            infiltrates and BD-
[alpha]       Cantarini                           related colitis
               et al.
               (2016)                        Complete remission of all
                             Case report      clinical manifestations
              Ideguchi
IVIg           et al.        Case series         Complete disease
               (2014)                              remission of
                                                 gastrointestinal,
             Lee et al.                           manifestations
               (2013)
                            Retrospective      Good response in two
              Kinoshita     observational     patients, remission in
               et al.           study        one, partial response in
               (2013)                          two, and unchanged GI
                                              lesions in two patients
               Hibi et
IFX          al. (2016)      Multicenter     IFX efficacy for patients
                           retrospective      with moderate-to-severe
              Tanida et         study              intestinal BD
             al. (2015)
                            Retrospective     Acceptable efficacy of
                            cohort study        IFX in BD patients
              Tanida et                            refractory to
             al. (2016)                       conventional treatments

                             Open-label         IFX efficacy in the
              Ma et al.         study         treatment of intestinal
               (2014)                                   BD

ADA                         Multicenter,       ADA effectiveness in
              Cantarini      open-label,     inducing and maintaining
               et al.       uncontrolled     clinical improvement and
               (2013)           study          remission in patients
                                                with intestinal BD
              Vitale et
             al. (2013)     Retrospective     Long-term efficacy and
                            observational      safety of ADA for the
              Deroux et         study         treatment of intestinal
             al. (2015)                         BD in the clinical
                                                      setting

                                               The relapse rate for
ETA                         Observational     etanercept therapy was
                                study        reduced significantly when
                                                   compared with
                                               conventional therapy

                                              Complete resolution of
ANA                          Case series       abdominal pain in two
                                                     patients,
                                              relapse in one patient

                             Case series      Complete resolution of
CANA                                              abdominal pain

TCZ                          Case series        Less effective for
                                             arthralgia and abdominal
                                                       pain

ADA, adalimumab; ANA, anakinra; anti-TNF-[alpha], anti-tumor necrosis
factor-[alpha]; 5-ASA, 5-aminosalicylic acid; AZA, azathioprine;
CANA, canakinumab; ETA, etanercept; INF-[alpha], interferon [alpha];
IFX, infliximab; IVIg, intravenous immunoglobulins; 6-MP,
6-mercaptopurine; MTX, methotrexate; SSZ, sulfasalazine; TCZ,
tocilizumab; THD, Thalidomide.
COPYRIGHT 2017 Hindawi Limited
No portion of this article can be reproduced without the express written permission from the copyright holder.
Copyright 2017 Gale, Cengage Learning. All rights reserved.

Article Details
Printer friendly Cite/link Email Feedback
Author:Lopalco, Giuseppe; Rigante, Donato; Venerito, Vincenzo; Fabiani, Claudia; Franceschini, Rossella; Ba
Publication:Mediators of Inflammation
Article Type:Report
Date:Jan 1, 2017
Words:8565
Previous Article:Chemokines (CCL3, CCL4, and CCL5) Inhibit ATP-Induced Release of IL-1[beta] by Monocytic Cells.
Next Article:Activated Platelets Induce an Anti-Inflammatory Response of Monocytes/Macrophages through Cross-Regulation of PG[E.sub.2] and Cytokines.
Topics:

Terms of use | Privacy policy | Copyright © 2024 Farlex, Inc. | Feedback | For webmasters |