uva-dare (digital academic repository) towards new ... fileserum biomarkers in...

1

Upload: dangtuong

Post on 16-Jun-2019

212 views

Category:

Documents


0 download

TRANSCRIPT

UvA-DARE is a service provided by the library of the University of Amsterdam (http://dare.uva.nl)

UvA-DARE (Digital Academic Repository)

Towards new measures of inflammation in spondyloarthritis

Turina, Maureen

Link to publication

Citation for published version (APA):Turina, M. C. (2016). Towards new measures of inflammation in spondyloarthritis.

General rightsIt is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s),other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons).

Disclaimer/Complaints regulationsIf you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, statingyour reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Askthe Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam,The Netherlands. You will be contacted as soon as possible.

Download date: 16 Jun 2019

Serum biomarkers in spondyloarthritis-

related diseases: lessons from psoriasis and

inflammatory bowel diseases

Maureen C. Turina, Robert Landewé, Dominique L. Baeten

Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and

immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam,

The Netherlands

Submitted for publication

2

20 SERUM BIOMARKERS IN PSORIASIS AND IBD

SUMMARY

Early diagnosis, monitoring of disease activity, prediction of treatment response, and structural outcome remain major challenges in spondyloarthritis (SpA). Biomarkers could play a role in addressing these challenges, but in SpA there is a lack of suitable biomarkers. As SpA is clinically and pathophysiologically closely related to psoriasis and inflammatory bowel disease (IBD), we reviewed the value of serum biomarkers in these conditions with the aim to find potential candidates for assessing SpA. Candidates of interest were antimicrobial peptides, including human beta defensin-2 (hBD-2) and lipocalin-2 (LCN-2), and class-1 MHC molecule beta2-microglobulin. Since these biomarkers are relevant in psoriasis and/or IBD from a pathophysiological point of view, and may play a role in the pathogenesis of SpA, we recommend further exploration of their value as biomarker in the diagnosis and prognosis of SpA.

21

2

CHAPTER 2

UNMET NEEDS IN SPONDYLOARTHRITIS

Spondyloarthritis (SpA) is an immune-mediated inflammatory disease affecting the spine, the peripheral joints and extra-articular tissues such as the skin, the gut, and the eye.1 The pathology of SpA is characterized by a combination of chronic tissue inflammation, destruction of cartilage and bone, and pathological new bone formation, that may lead to ankylosis. Despite major advances in the field, such as the use of magnetic resonance imaging (MRI) to detect spinal inflammation and to find appropriate patients for treatment with TNF blockers,2–8 several important challenges remain to be addressed.

First, there is a huge diagnostic delay of approximately 5-10 years between the onset of SpA symptoms and the diagnosis.9–11 This delay is due to the fact that the early clinical signs and symptoms are often non-specific, that pathognomonic features of SpA, such as radiologic sacroiliitis, only appear late in the disease course, and that well-known biological markers such as HLA-B27 lack specificity (many healthy individuals carry HLA-B27) and therefore predictive value in the diagnostic context. Second, good biological predictors for treatment response are lacking. Currently, the only informative marker is C-reactive protein (CRP). Nevertheless, CRP as a marker for treatment response falls short because only one third of the patients have elevated CRP levels prior to treatment. Third, the progression rate of new bone formation as observed on consecutive X-rays of the lumbar and cervical spine is very slow and besides only occurs in a relatively small group of patients. Currently, markers predicting which patients will develop significant new bone formation are lacking.

Overall, an early diagnosis, a proper prediction of structural damage, as well as prediction of treatment response, all in individual patients, remain major challenges in SpA. Biomarkers could be of value in addressing these challenges.

BIOMARKERS IN SPONDYLOARTHRITIS

A biomarker is a “characteristic that can be objectively measured and evaluated as an indicator of a normal biologic process, a pathophysiologic process, or a pharmacologic response to a therapeutic intervention”.12 A biomarker should be sensitive, specific, reproducible, and deriving the biomarker from a patient should preferably be a non-invasive procedure. Taking these criteria into account, biomarkers can have an important role to fulfill the previously defined unmet needs in the process of diagnosis, treatment response and prognosis. Several biomarkers have already been studied in SpA. Common markers of systemic inflammation such as CRP and erythrocyte sedimentation rate (ESR) have been used for a long time in the clinical care of SpA patients. Accordingly, elevated CRP is included as a criterion in the ASAS classification criteria for axial SpA criteria but not for ASAS peripheral SpA criteria.2,13

22 SERUM BIOMARKERS IN PSORIASIS AND IBD

When elevated, CRP levels may correlate with disease activity measures and decrease upon effective treatment.14–17 Moreover, CRP appears to be a predictor for radiographic axial progression.18 However, CRP and ESR are only elevated in 30 to 50% of the SpA patients with clinical signs and symptoms of active disease and inflammation14–17 and, obviously, are not specific for SpA. The limited sensitivity and specificity explains why CRP and ESR can be useful biomarkers at the group level (such as in clinical trials) but lack sufficient predictive value to be used in individual patients.

Other serum markers of inflammation that have been studied in SpA are Interleukin-6 (IL-6) and calprotectin. IL-6 is the main driver of CRP and, similar to CRP, serum IL-6 levels are significantly increased in active SpA and decrease upon clinical response after tumour necrosis factor (TNF) blockade.19,20 The calcium-binding protein calprotectin, a heterodimer of S100A8 and S100A9, is expressed and secreted during monocyte infiltration into inflamed tissues, including macrophage infiltration in synovial tissue of SpA patients.21,22 Calprotectin serum levels are elevated in SpA, correlate moderately well (r=0.40-0.60) with disease activity, and decrease upon TNF blockade.21–23 Moreover, calprotectin serum levels is an independent predictor for radiological axial progression at the group level.24 Two other biomarkers that were studied in SpA are more related to the pathological processes than to inflammation as such. Vascular endothelial growth factor (VEGF), a growth factor involved in neo-angiogenesis, may be involved in the pronounced hypervascularity in SpA synovitis.25,26 Serum VEGF levels are significantly elevated in SpA versus healthy controls and significantly albeit moderately at best correlate (r=0.22-0.44) with disease activity, but levels do not decrease after short-term treatment with TNF blockers.20,27 Whereas an original report indicated that serum VEGF levels were predictive of new bone formation,28 these results could not be confirmed by others.29

Matrix metalloproteinase-3 (MMP-3) is an enzyme involved in tissue remodelling and cartilage damage. MMP-3 expression was elevated in synovial tissue, synovial fluid and serum in SpA and this expression correlated well with disease activity30–34 and normalized upon effective treatment.30

With regard to predicting new bone formation in ankylosing spondylitis (AS), the prototype of axial SpA, most studies have focused on molecules involved in the regulation of osteoblast activity, including the wnt-pathway, which is involved in the process of new bone formation. Dickkopf-1 (DKK1) and sclerostin are inhibitors of this pathway, and Diarra et al.35 found that serum DKK1 levels were decreased in AS patients versus healthy controls or rheumatoid arthritis patients and low levels were associated to radiographic axial progression as expressed by syndesmophytes formation. These results were confirmed by others.36–38 For sclerostin, discordant results were seen regarding the relationship with radiographic axial spinal progression.36,39–42

Finally, several studies have screened for disease-specific autoantibodies as potential

23

2

CHAPTER 2

diagnostic biomarker in SpA. Unfortunately, most studies did not find disease-specific autoantibodies and the few that were reported could not be independently confirmed.43–45 Recently, one group found elevated levels of IgG autoantibodies CD74 specific for HLA class II-associated invariant chain peptide (CLIP) in SpA.46,47 These data as well as the diagnostic value of this autoantibody should be validated in independent cohorts.

In summary, despite major efforts over the last years only few biomarkers have been described and validated in SpA, and often these biomarkers are useful at the group level but lack specificity to be used in individual patients in clinical care. Interestingly, SpA is genetically, pathophysiologically and clinically associated with psoriasis and inflammatory bowel diseases (IBD), two conditions in which serum biomarkers have been described. Therefore, we reviewed here the value of biomarkers in these conditions and discuss their potential utility for SpA. Reviewing was done for the settings of diagnosis (i.e. difference between disease and healthy or disease and similar inflammatory diseases), with respect to correlation with disease activity and response to effective treatment, as well as for predicting structural damage (in axial SpA). We have evaluated both the pathophysiological and clinical value of these markers.

SERUM BIOMARKERS IN PSORIASIS

Psoriasis is an immune-mediated, chronic inflammatory disease primarily of the skin but can also affect nails and peripheral and axial joints. The disease is characterized by abnormal differentiation and hyperproliferation of epidermal keratinocytes and infiltration of immune cells. Roughly 1 in 5 psoriasis patients will develop psoriatic arthritis (PsA), one of the phenotypical forms of SpA.48 At the genetic level, SNPs in IL-23R and Th-17 genes have shown to be associated with both psoriasis and SpA.

In clinical practice, a diagnosis of psoriasis is based on clinical history and examination and, in case of diagnostic uncertainty, skin biopsies. Disease activity is mainly determined by the Psoriasis Area and Severity Index (PASI),49 which includes the percentage of affected skin area, the erythema, the induration, and scaling. Currently, serum biomarkers do not play an important role in clinical practice but are extensively studied in clinical trials (Table 1).

Serum CRP levels were found to be elevated in the moderate and severe forms of psoriasis but not in patients with mild disease, when compared with the healthy individuals.50,51 Several studies have reported CRP elevation in 17-45.7% of the psoriasis patients versus 1% in the control group.51–53 Furthermore, CRP levels decreased significantly after effective treatment was started.51,54–57 It is therefore only a potentially useful marker for disease activity in patients who did not receive systemic treatment for at least one month. In the treated patients, PASI is preferred to monitor disease activity. ESR levels were elevated in

24 SERUM BIOMARKERS IN PSORIASIS AND IBD

Table 1. Serum biomarkers in psoriasis: elevation in comparison with healthy controls, correlation with disease activity according to PASI, and modulation by effective treatment

Biomarker

Elevation incomparison with healthy controls

Correlation withdisease activitymeasured PASI Treatment response References

CRP + +/- +/- [51–55]ESR + NA NA [58]TNFα + NA + [59, 60]TNFα-R1 + +/- NA [59, 66, 77, 99]IFNγ + NA NA [60]IL-8 + - NA [59, 60, 63]IL-12 +/- + NA [60]IL-17 +/- + + [59, 60, 64, 65]IL-18 + + + [60, 61, 70]IL-22 + + + [59, 62, 65]E-selectin + +/- NA [63, 66–68]ICAM-1 + + +/- [63, 66, 67, 69, 70]VAP-1 + - - [72–74]S100A8/A9 (calprotectin) + + NA [75, 76]LCN-2 + +/- - [77–80]hBD-2 + + +/- [81, 82]Resistin + +/- +/- [83, 90, 91]Leptin + - - [83–87, 90]Adiponectin - +/- +/- [84–86, 88, 90, 92]Ghrelin + + - [89, 90]RBP-4 + + - [80, 85]VEGF + + +/- [59, 62, 93–95]Nitric oxide + +/- + [94, 96–98]

CRP, C-reactive protein; ESR, erythrocyte sedimentation rate; hBD-2, human beta-defensin-2; ICAM-1, intracellular adhesion molecule-1; IFNγ, interferon gamma; IL, interleukin; LCN-2, lipocalin-2; NA, not applicable; PASI, Psoriasis Area and Severity Index; RBP-4, retinol binding protein-4; TNF, tumor necrosis factor; TNFα-R1, tumor necrosis factor alpha-receptor 1; VEGF, vascular endothelial growth factor; VAP-1, vascular adhesion protein-1.

approximately half of the psoriasis patients compared to healthy controls. Elevated ESR was clearly more frequent in the active state of the disease (80%) versus non-active psoriasis.58

Cytokines, including interferon-gamma (IFNγ), TNF-α, TNF-R1, IL-6, IL-8, IL-12, and IL-18, IL-22, and IL-23 have been investigated thoroughly in psoriasis. Coimbra et al. and others59–63 have found that the serum levels of these cytokines were overall significantly higher in psoriasis as compared with healthy controls. But the data on the correlation with PASI were rather conflicting. Psoriasis-specific treatment including methotrexate, UV-A/UV-B treatment, and TNF blockade did have an effect on serum levels of these cytokines.59,60,64,65

Adhesion molecules are expressed by activated tissue cells (e.g. endothelium) as well as leukocytes and allow selective migration of distinct leukocyte subsets to specific tissues and

25

2

CHAPTER 2

sites of inflammation. For example, activated epithelial cells express E-selectin, which can interact with E-selectin on another epithelial cell or with alpha E beta 7 (a member of the integrin family) on CD8 positive T cells. Of the soluble adhesion molecules that can easily be measured in serum, E-selectin, ICAM-1, and VAP-1 are best studied in psoriasis.63,66–74 All of these molecules showed elevated serum levels in psoriasis versus healthy controls. Soluble E-selectin and ICAM-1 levels, however, were consistently (but only weakly to moderately) associated with disease activity as expressed by PASI scores (r=0.44 and r=0.26-0.43, respectively). Accordingly, only E-selectin levels decreased significantly and consistently upon effective treatment.63,67,68,71,72

Among the antimicrobial peptides and proteins (AMP), calprotectin (S100A8/A9 proteins), lipocalin-2 (LCN-2, also called neutrophil gelatinase-associated lipocalin [NGAL]), and human β-defensin-2 (hBD-2) were associated with psoriasis. Highly up-regulated calprotectin levels were found in a proteomic analysis of the epidermis from psoriasis patients.75 In addition, Benoit et al.76 showed that calprotectin levels in serum were significantly elevated when compared to healthy controls and that patients with a high PASI (>15) score also had higher levels of calprotectin. Regarding LCN-2 serum levels, several groups consistently found elevated levels in psoriasis patients when compared with healthy individuals. However, absent or only weak (r=0.27) correlations were found between LCN-2 levels and PASI scores, and serum levels were not normalized by effective treatment.77–80 Serum levels of another AMP, hBD-2, were elevated consistently when compared with healthy donors. Jansen et al.81 reported a high correlation with PASI (r=0.82), whereas another group did not see such a strong correlation (r<0.1, p>0.05).82

Adipokines are produced by fat (adipose) tissue and play a role in metabolic processes, but these proteins are also associated with inflammation. Extensively studied adipokines were resistin, leptin, adiponectin, ghrelin, and retinol binding protein-4 (RBP-4). With the exception of adiponectin, all serum and plasma adipokine levels were significantly higher in psoriasis versus healthy individuals.80,83–89 Only resistin levels correlated weakly with PASI (r2=0.027) and none of the adipokines consistently showed a normalization in levels after effective treatment.80,85,88,90–92

Growth factors are involved in angiogenesis and hypervascularity in psoriasis and are well studied as biomarkers. Of these growth factors, VEGF was the most evaluated markers. VEGF was the most promising in psoriasis, since there were differences in serum levels between psoriasis patients and healthy controls and there was a strong correlation (R=0.65) with PASI.59,93,94 However, serum levels did not normalize upon effective treatment.59,62,95

Nitric oxide (NO) is a free radical and is secreted by various cell types, including fibroblasts and endothelial cells. Tekin et al.96 performed a study in 22 psoriasis samples versus 21 healthy controls and found significantly higher serum levels in the former group. These results were reproduced by others.94,97,98 Furthermore, NO plasma and serum levels

26 SERUM BIOMARKERS IN PSORIASIS AND IBD

decreased after effective treatment.96

In summary, CRP, ESR, IL-18, IL-22, ICAM-1, calprotectin, LCN-2, hBD-2, VEGF, RBP-4, and NO were the most informative biomarkers since these markers are elevated in patients with the disease when compared to healthy controls, show weak to moderate correlations with disease activity, and/or decrease upon effective treatment at the group level, indicating that -at least from a pathophysiological point of view- these are interesting markers. In spite of this, though, the value of these markers as biomarker in clinical practice (at the individual patient level) seems –if investigated- modest at best. In psoriasis, CRP and ESR were the only serum biomarkers used in clinical practice for the evaluation of treatment, despite the disappointing results earlier mentioned. The most interesting serum and plasma biomarkers in psoriasis from a pathophysiological point of view are the antimicrobial peptides hBD-2 and LCN-2: the expression of both peptides is driven by IL-17 and may therefore reflect a key pathophysiological process in psoriasis skin, although the role in clinical practice remains inconclusive.

SERUM BIOMARKERS IN IBD

IBD includes Crohn’s disease (CD) and ulcerative colitis (UC). Peripheral arthritis and sacroiliitis are seen in 7-16%100–102 and 2-32%100,102,103 of the IBD patients, respectively. Similarly, 60-70% of the SpA patients show subclinical signs of gut inflammation on biopsies obtained by ileocolonscopy,104,105 but only 7% of these patients will eventually develop clinically overt IBD over time. The link between SpA and IBD is corroborated by genetic studies, with the best example being polymorphisms in the interleukin 23 receptor (IL23R) gene which are associated both with IBD (OR 0.38 and 0.73 for CD and UC, respectively) and AS (OR=0.53-1.27).106–109

The diagnosis of IBD in clinical practice is based on the combination of clinical history, physical examination, stool findings (i.e. calprotectin and lactoferrin), serological antibodies, inflammatory markers in blood, and colonoscopy.110 The major clinical outcome measure in Crohn’s disease, the Crohn’s Disease Activity Index (CDAI),111 includes clinical and laboratory findings to determine disease activity. In UC, several scores are used to determine disease activity including clinical and endoscopic scores. A frequently used clinical score is Simple Clinical Colitis Activity Index (SCCAI).112 The Mayo activity index is based on clinical as well as endoscopic findings.113

Of the acute phase reactants, CRP was found to be a biomarker for IBD since the serum levels were elevated in CD and to a lesser extent in UC when compared with healthy controls or functional bowel disorders.114,115 Elevated levels were correlated with a higher disease activity.116–118 Louis et al.118 studied a group of 226 CD patients receiving TNF-inhibitors and

27

2

CHAPTER 2

a clinical response was associated with a decrease in CRP with similar results in a larger cohort. There were discrepant data about the ability of CRP to predict disease relapse.119,120 Other well studied inflammatory markers were ESR, procalcitonin, and YKL-40. All three markers correlated with the disease activity but only ESR and procalcitonin differentiated between IBD and healthy individuals or other gastrointestinal diseases.121–128 Moreover, a trend towards elevated levels of YKL-40 was found in more severe CD patients having strictures (i.e. narrowing of the intestinal lumina),124 but since the majority of active patients do not have elevated levels, in clinical practice YKL-40 is of little value.

Cytokines measured in serum did not seem to be important markers of disease in IBD, despite their role in the inflammatory process.129,130 The only potential marker is IL-6, the main driver of CRP. Levels of IL-6 in serum were increased in active CD compared with healthy controls but correlation with disease activity was weak.131–133

Of the soluble adhesion molecules, ICAM-1 was the most promising serum and plasma marker as the levels were elevated when compared with healthy controls. Although discrepant data are available on correlation with disease activity, effective treatment down-regulates ICAM-1 levels.134–138 E-selectin levels in serum or plasma were significantly higher in IBD than in healthy controls. However, there was no significant association between disease activity and treatment.134–138 A few studies were published on vascular cell adhesion molecule (VCAM). The serum or plasma levels in IBD were not consistently elevated versus healthy controls and correlations to disease activity were poor.134–136

The adipokines adiponectin, leptin, ghrelin, resistin, and vaspin had been investigated thoroughly in IBD. Only serum levels of resistin and vaspin were consistently elevated in IBD versus healthy individuals. Moreover, strong correlations with PASI were found for ghrelin, resistin and vaspin levels with disease activity.139–147 Ghrelin and resistin levels were decreased upon effective treatment but this was not studied for the other adipokines.148,149

Of the S100 proteins, S100A12 levels in serum were elevated in IBD when compared with healthy controls, but there are conflicting data about the correlation with active disease activity.150,151 Serum levels of S100A8/S100A9, calprotectin, were elevated in IBD versus healthy individuals and correlated with the CDAI.152–154 Similar results were seen in children using pediatric CDAI indices.155 Furthermore, calprotectin serum levels decreased upon treatment with TNF inhibition.154,156 Fecal calprotectin is one of the most extensively studied stool markers in IBD. Calprotectin levels in stool can differentiate between IBD and healthy controls or irritable bowel syndrome (IBS). One meta-analysis of Von Roon et al.157 included 30 studies with 1210 IBD patients, 297 colorectal cancer, 697 IBS, and 3393 healthy controls and reported that fecal calprotectin is a useful biomarker to differentiate between IBD and non-IBD patients. The overall weighted area under curve (AUC) was with 0.95-0.98 (using a cut-off level of 50 ug/g and 100ug/g, respectively) exceptionally high. Moreover, differentiating CD from IBS by fecal calprotectin showed an AUC of 0.97. Van Rheenen et

28 SERUM BIOMARKERS IN PSORIASIS AND IBD

al.158 determined that fecal calprotectin could be used as screening tool in suspected clinical IBD, thereby decreasing the use of invasive colonoscopy by 67%, but at the cost of false negative results in 6% of the cases. Results of a meta-analysis that included 13 studies (1471 IBD patients), showed that fecal calprotectin is also a good marker for disease activity based on the Mayo activity index.159 Moreover, fecal calprotectin decreased upon treatment160–163 and may predict disease relapse.120,164,165

Several AMPs have been investigated in IBD. Serum LCN-2 (NGAL) levels were elevated in IBD when compared with healthy controls and IBS.166–168 HBD-2 seemed a potential marker since expression levels were increased in inflamed mucosa versus non-inflamed mucosa of the colon.169 However, Yamaguchi et al. found no differences in hBD-2 plasma levels in UC or CD when compared to healthy controls.170

Beta-2-microglobulin (β2-M), which forms a part of the major histocompatibility complex class 1 (MHC 1), had been investigated in two independent studies. In 2001 Zissis et al.171 showed that serum levels were elevated in CD versus healthy controls, and that serum levels were higher when patients had a higher disease activity, but they did not see similar results for UC. Yilmaz et al.172 recently showed similar results but now in the whole IBD population.

Of the serological antibodies studied in IBD,173–177 anti-saccharomyces cervisiae antibodies (ASCA) and perinuclear anti-neutrophil cytoplasmic antibodies (pANCA) showed the highest diagnostic accuracy. Sensitivity and specificity for ASCA were 60% and 91% and for pANCA 50% and 95% respectively, when distinguishing IBD from healthy controls. Sensitivity and specificity for distinguishing between CD and UC were for ASCA (CD): 60% and 86%, and for pANCA (UC): 50% and 94% respectively.178 Similar results were seen in other studies.179,180 Also, when discriminating CD from UC or when comparing with other gastrointestinal diseases, including celiac disease, the specificity was too low for these antibodies.174–178 Both autoantibodies had a positive predictive value of 80-90% and a negative predictive value of 50-80%. The number of false-positive and false-negative individual was too high to recommend these autoantibodies for routine diagnostic procedures. Ester et al.181 found no association of ASCA or pANCA with response to infliximab treatment, but ASCA positivity was associated with worse disease progression: a more severe disease type (from inflammation to structuring (or stenosis) and penetrating) disease in CD.182 Recently, Bonneau et al.183 published a systematic review in which anti-glycan, anti-GP2, and anti-GM-CSF antibodies were also described as having low sensitivity but high specificity to CD when compared with UC but the contribution of these antibodies to diagnosis is not yet clear.

To summarize, CRP and ESR, the S100 family proteins S100A12 and S100A8/A9 (calprotectin), the antimicrobial peptides LCN-2 and hBD-2, the MHC class 1 molecule β2-M, and ASCA antibodies are of interest as biomarkers in IBD from a pathophysiological point of view. In clinical practice however, only a few of these markers are used, including CRP, ESR, and fecal calprotectin. Since CRP correlates with disease activity and CRP levels normalize upon

29

2

CHAPTER 2

effective treatment, evaluation of therapy by CRP is often applied in general practice. Fecal calprotectin, but not serum calprotectin, is extensively used in clinical practice to identify IBD patients with intestinal inflammation, providing high sensitivity and specificity in detecting IBD. Nevertheless, the ileocolonoscopy remains the gold standard. With regard to antibodies, ASCA antibodies are able to differentiate between CD and UC and are used in clinical practice although not as a screening diagnostic procedure. Other markers in IBD that are of pathophysiological interest are the IL-17 driven LCN-2, hBD-2, and β2-M, that forms part of the MHC class-I molecule (Table 2).

Table 2. Serum biomarkers in IBD levels: elevation in comparison with healthy controls, correlation with disease activity according to CDAI, SCCAI, or endoscopic activity, modulation by effective treatment, and outcome of structural damage

Biomarker

Elevation In comparison with healthy controls

Correlation with disease activity measured by CDAI or SCCAI or endoscopic activity

Treatment response

Outcome structural damage References

CRP + + + + [114–121]ESR + + NA NA [114, 128]Procalcitonin + +/- NA NA [125–127]YKL-40 +/- + NA +/- [122–124]IL-6 + +/- NA NA [131–133, 156]ICAM-1 +/- - + NA [134–138]E-selectin + - - NA [134–138]VCAM +/- - - NA [134–136]Adiponectin + - - NA [145–147, 149, 184]Leptin +/- - +/- NA [145, 146, 148, 149, 184]Ghrelin + + + +/- [140, 141, 146, 184, 185]Resistin + +/- + NA [142, 143, 146, 149, 184]Vaspin +/- - NA NA [144]S100A12 + +/- + NA [150, 151]S100A8/A9 + NA NA + [152–155]LCN-2 + + NA NA [166–168]hBD-2 - - NA NA [169, 170]β2-M + + NA NA [171, 172]

ASCA + + - +[173, 175, 177, 178, 181, 182]

pANCA + - + - [173–178, 181, 182]

ASCA, anti-saccharomyces cerevisiae antibodies; β2-M, β2-microglobulin; CDAI, crohn’s disease activity index; CRP, C-reactive protein; ESR, erythrocyte sedimentation rate; hBD-2, human beta-defensin-2; ICAM, intracellular adhesion molecule-1; IL, interleukin; LCN-2, lipocalin-2; NA, not applicable; pANCA, perinuclear anti-neutrophil cytoplasmic antibodies; SCCAI, simple clinical colitis activity index; VCAM, vascular cell adhesion molecule; VEGF, vascular endothelial growth factor.

30 SERUM BIOMARKERS IN PSORIASIS AND IBD

LESSONS TO BE LEARNED FROM POTENTIAL SERUM

BIOMARKERS IN PSORIASIS AND IBD

Several biomarkers studied in psoriasis and IBD may be informative in SpA as well. CRP, ESR, calprotectin, and VEGF are interesting in psoriasis and IBD. These markers have already been studied in SpA (as mentioned above). While they may be discriminatory at the group level, sensitivity and/or specificity are too low to be used in clinical practice for diagnosis, disease activity measurement, evaluation of treatment response, or prediction of axial spinal progression.

Some of the biomarkers of interest in in psoriasis and IBD have also been studied in SpA. Negative or variable results were found for serum-IL-18,186 ICAM-1,187–189 RBP-4,190,191 and NO.192,193

ASCA antibodies can help to discriminate CD from UC, have already been studied in SpA. The levels of ASCA antibodies were found to be higher in SpA patients than in healthy controls, as reported by different studies.194,195 One study determined these antibodies in non-IBD SpA patients, and 30% of them appeared to be ASCA-positive. Nevertheless, levels did not correlate with disease activity.196

The AMPs are a new family of relevant biomarkers in psoriasis and IBD as was found in several studies. AMPs have not yet been extensively studied in SpA. The most interesting members of this family are LCN-2 and hBD-2, both of which are driven by IL-17 and are reflecting active disease in psoriasis as well as IBD. And because the reliable measurement of IL-17 in serum remains a challenge, these biomarkers may be used as a proxy for IL-17-mediated tissue inflammation in psoriasis and IBD. As SpA is also an IL-17-driven disease,197–201 these markers certainly deserve further study in this condition. It should be noted, however, that these AMPs are mainly produced by epithelial cells and may thus be more useful in skin and gut diseases than in other IL-17-related inflammatory conditions. Along the same line, IL-22 is an IL-10 cytokine family member which is produced by Th17 cells under the control of IL-23. It was shown that plasma IL-22 levels were increased in SpA versus healthy individuals, but no correlation was found with Bath ankylosing spondylitis disease activity index (BASDAI).202 The reliability of serum IL-22 measurement and the validity of these findings require replication in independent studies.

Another biomarker which was identified in IBD and may be directly related to the pathological processes in SpA is β2-M. β2-M is a component of class I MHC molecules, including HLA-B27, which is (still) the major genetic risk factor for SpA. Although the exact role of HLA-B27 in the disease pathogenesis is not yet clear, several hypotheses suggest that the level of expression of β2-M may modulate the pathological role of HLA-B27. One hypothesis proposes that β2-

31

2

CHAPTER 2

M-free HLA-B27 heavy chains form disulphide-linked homodimers which can be recognized by the KIR3DL2 receptors on NK-cells and T-cells.203 The second hypothesis concerns HLA-B27 heavy chain misfolding prior to binding of β2-M and peptide, resulting in unfolded protein responses (UPR), which subsequently may lead to an inflammatory cytokine release.201,204 Interestingly, modulation of β2-M expression levels in the HLA-B27 transgenic rat model of SpA did profoundly affect the phenotype of the disease,205–207 confirming the potential importance of this molecule in SpA. Therefore, serum β2-M expression levels should be further explored as potential biomarker in SpA.

EXPERT COMMENTARY

To conclude, of the serum biomarkers identified in psoriasis and IBD, the IL-17 driven anti-microbial peptides hBD-2 and LCN-2, and the class I MHC molecule β2-microglobulin are the most interesting candidates from a pathophysiological standpoint to be further explored in SpA for clinical purposes.

KEY POINTS

• SpA is an immune-mediated inflammatory disease affecting the spine, the peripheral joints, and extra-articular tissues such as the skin, the gut, and the eye.

• Serum biomarkers could address several challenges in SpA (diagnosis, disease activity monitoring, treatment response, and outcome).

• Serum biomarkers well studied in spondyloarthritis are CRP, ESR, IL-6, calprotectin, VEGF, MMP-3, DKK-1, and sclerostin.

• Although the aforementioned biomarkers are useful at the group level in SpA, these markers lack specificity for the use in individual patients in clinical care.

• SpA is a clinically and pathophysiologically closely related to psoriasis and inflammatory bowel disease.

• In psoriasis, CRP, ESR, IL-18, IL-22, ICAM-1, calprotectin, LCN-2, hBD-2, VEGF, RBP-4, and NO were the most interesting markers from a pathophysiological standpoint.

• In IBD, CRP and ESR, hBD-2, LCN-2, and ASCA antibodies were of interest from a pathophysiological point of view.

• HBD-2, LCN-2, and β2-microglobulin are the most interesting candidates from a pathophysiological standpoint to be further explored in SpA for clinical purposes.

32 SERUM BIOMARKERS IN PSORIASIS AND IBD

REFERENCES

1. Dougados M, Baeten D: Spondyloarthritis. Lancet 2011, 377:2127–37.

2. Sieper J, Rudwaleit M, Baraliakos X, Brandt J, Braun J, Burgos-Vargas R, Dougados M, Hermann K-G, Landewé R, Maksymowych W, van der Heijde D: The Assessment of SpondyloArthritis international Society (ASAS) handbook: a guide to assess spondyloarthritis. Ann Rheum Dis 2009, 68 Suppl 2:ii1–44.

3. Van Den Bosch F, Kruithof E, Baeten D, Herssens A, de Keyser F, Mielants H, Veys EM: Randomized double-blind comparison of chimeric monoclonal antibody to tumor necrosis factor alpha (infliximab) versus placebo in active spondylarthropathy. Arthritis Rheum 2002, 46:755–65.

4. Braun J, Brandt J, Listing J, Zink a, Alten R, Golder W, Gromnica-lhle E, Kellner H, Krause a, Schneider M, Sörensen H, Zeidler H, Thriene W, Sieper J: Treatment of active ankylosing spondylitis with infliximab: a randomised controlled multicentre trial. Lancet 2002, 359:1187–1193.

5. Mease PJ, Goffe BS, Metz J, VanderStoep A, Finck B, Burge DJ: Etanercept in the treatment of psoriatic arthritis and psoriasis: a randomised trial. Lancet 2000, 356:385–390.

6. Paramarta JE, De Rycke L, Heijda TF, Ambarus C a, Vos K, Dinant HJ, Tak PP, Baeten DL: Efficacy and safety of adalimumab for the treatment of peripheral arthritis in spondyloarthritis patients without ankylosing spondylitis or psoriatic arthritis. Ann Rheum Dis 2013, 72:1793–9.

7. Sieper J, van der Heijde D, Dougados M, Mease PJ, Maksymowych WP, Brown M a, Arora V, Pangan AL: Efficacy and safety of adalimumab in patients with non-radiographic axial spondyloarthritis: results of a randomised placebo-controlled trial (ABILITY-1). Ann Rheum Dis 2013, 72:815–22.

8. Landewé R, Braun J, Deodhar a, Dougados M, Maksymowych WP, Mease PJ, Reveille JD, Rudwaleit M, van der Heijde D, Stach C, Hoepken B, Fichtner a, Coteur G, de Longueville M, Sieper J: 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.

9. Feldtkeller E, Khan MA, van der Heijde D, van der Linden S, Braun J: Age at disease onset and diagnosis delay in HLA-B27 negative vs. positive patients with ankylosing spondylitis. Rheumatol Int 2003, 23:61–6.

10. Salvadorini, F. Bandinelli, A. Delle Sedie, L. Riente, A. Candelieri, S. Generini, N. Possemato, S. Bombardieri MM-C: Ankylosing spondylitis: how diagnostic and therapeutic delay have changed over the last six decades. Clin Exp Rheumatol 2012, 30:561–565.

11. Sorensen J, Hetland ML: Decreases in diagnostic delay are supported by sensitivity analyses. Ann Rheum Dis

2014, 73:e45–e45.

12. Gruttola VG De, Clax P, Demets DL, Downing GJ, Ellenberg SS, Friedman L, Gail MH, Prentice R, Wittes J, Zeger SL: Considerations in the Evaluation of Surrogate Endpoints in Clinical Trials: Summary of a National Institutes of Health Workshop. Control Clin Trials 2001, 1998:485–502.

13. Rudwaleit M, van der Heijde D, Landewé R, Akkoc N, Brandt J, Chou CT, Dougados M, Huang F, Gu J, Kirazli Y, Van den Bosch F, Olivieri I, Roussou E, Scarpato S, Sørensen IJ, Valle-Oñate R, Weber U, Wei J, Sieper J: The Assessment of SpondyloArthritis International Society classification criteria for peripheral spondyloarthritis and for spondyloarthritis in general. Ann Rheum Dis 2011, 70:25–31.

14. 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 (Oxford) 2010, 49:536–41.

15. Poddubnyy D a, Rudwaleit M, Listing J, Braun J, Sieper J: Comparison of a high sensitivity and standard C reactive protein measurement in patients with ankylosing spondylitis and non-radiographic axial spondyloarthritis. Ann Rheum Dis 2010, 69:1338–41.

16. Visvanathan S, Wagner C, Marini JC, Baker D, Gathany T, Han J, van der Heijde D, Braun J: Inflammatory biomarkers, disease activity and spinal disease measures in patients with ankylosing spondylitis after treatment with infliximab. Ann Rheum Dis 2008, 67:511–7.

17. De Vries MK, van Eijk IC, van der Horst-Bruinsma IE, Peters MJL, Nurmohamed MT, Dijkmans B a C, Hazenberg BPC, Wolbink GJ: Erythrocyte sedimentation rate, C-reactive protein level, and serum amyloid a protein for patient selection and monitoring of anti-tumor necrosis factor treatment in ankylosing spondylitis. Arthritis Rheum 2009, 61:1484–90.

18. Poddubnyy D, Rudwaleit M, Haibel H, Listing J, Märker-Hermann E, Zeidler H, Braun J, Sieper J: Rates and predictors of radiographic sacroiliitis progression over 2 years in patients with axial spondyloarthritis. Ann Rheum Dis 2011, 70:1369–74.

19. Gratacós J, Collado A, Filella F, Sanmartí R, Cañete J, Llena J, Molina R, Ballesta A, Muñoz-Gómez J: Serum cytokines (IL-6, TNF-alpha, IL-1 beta and IFN-gamma) in ankylosing spondylitis: a close correlation between serum IL-6 and disease activity and severity. Br J Rheumatol 1994, 33:927–931.

20. Pedersen SJ, Hetland ML, Sørensen IJ, Ostergaard M, Nielsen HJ, Johansen JS: Circulating levels of interleukin-6, vascular endothelial growth factor, YKL-40, matrix metalloproteinase-3, and total aggrecan in spondyloarthritis patients during 3 years of treatment with TNFα inhibitors. Clin Rheumatol 2010, 29:1301–

33

2

CHAPTER 2

9.

21. Kane D, Roth J, Frosch M, Vogl T, Bresnihan B, FitzGerald O: Increased perivascular synovial membrane expression of myeloid-related proteins in psoriatic arthritis. Arthritis Rheum 2003, 48:1676–85.

22. Kruithof E, De Rycke L, Vandooren B, De Keyser F, FitzGerald O, McInnes I, Tak PP, Bresnihan B, Veys EM, Baeten D: Identification of synovial biomarkers of response to experimental treatment in early-phase clinical trials in spondylarthritis. Arthritis Rheum 2006, 54:1795–804.

23. De Rycke L, Baeten D, Foell D, Kruithof E, Veys EM, Roth J, De Keyser F: Differential expression and response to anti-TNFalpha treatment of infiltrating versus resident tissue macrophage subsets in autoimmune arthritis. J Pathol 2005, 206:17–27.

24. Turina MC, Sieper J, Yeremenko N, Conrad K, Haibel H, Rudwaleit M, Baeten D, Poddubnyy D: Calprotectin serum level is an independent marker for radiographic spinal progression in axial spondyloarthritis. Ann Rheum Dis 2014, 73:1746–8.

25. Baeten DL, Demetter P, Cuvelier C, Bosch F Van den, Kruithof E, Damme N van, Verbruggen G, Mielants H, Veys E, Keyser F De: Comparative study of the synovial histology in rheumatoid arthritis , spondyloarthropathy , and osteoarthritis : influence of disease duration and activity. Ann Rheum Dis 2000, 59:945–953.

26. Baeten D, Kruithof E, De Rycke L, Vandooren B, Wyns B, Boullart L, Hoffman IE a, Boots AM, Veys EM, De Keyser F: Diagnostic classification of spondylarthropathy and rheumatoid arthritis by synovial histopathology: a prospective study in 154 consecutive patients. Arthritis Rheum 2004, 50:2931–41.

27. Drouart M, Saas P, Billot M, Cedoz J-P, Tiberghien P, Wendling D, Toussirot E: High serum vascular endothelial growth factor correlates with disease activity of spondylarthropathies. Clin Exp Immunol 2003, 132:158–62.

28. Poddubnyy D, Conrad K, Haibel H, Syrbe U, Appel H, Braun J, Rudwaleit M, Sieper J: Elevated serum level of the vascular endothelial growth factor predicts radiographic spinal progression in patients with axial spondyloarthritis. Ann Rheum Dis 2014, 73:2137–43.

29. Baraliakos X, Hermann KG, Xu S, Hsu B, Braun J: VEGF and CRP serum levels lack predictive value for outcomes as assessed by conventional radiographs and magnetic resonance imaging in patients with active ankylosing spondylitis treated with the TNF inhibitor golimumab. Ann Rheum Dis 2014;73(Suppl2):1138-1139.

30. Vandooren B, Kruithof E, Yu DTY, Rihl M, Gu J, De Rycke L, Van Den Bosch F, Veys EM, De Keyser F, Baeten D: Involvement of matrix metalloproteinases and their inhibitors in peripheral synovitis and down-regulation by tumor necrosis factor alpha blockade in spondylarthropathy. Arthritis Rheum 2004, 50:2942–53.

31. Chen C-H, Lin K-C, Yu DTY, Yang C, Huang F, Chen H, Liang T-H, Liao H-T, Tsai C-Y, Wei JCC, Chou C-T: Serum

matrix metalloproteinases and tissue inhibitors of metalloproteinases in ankylosing spondylitis: MMP-3 is a reproducibly sensitive and specific biomarker of disease activity. Rheumatology (Oxford) 2006, 45:414–20.

32. Yang C, Gu J, Rihl M, Baeten D, Huang F, Zhao M, Zhang H, Maksymowych WP, De Keyser F, Veys EM, Yu DTY: Serum levels of matrix metalloproteinase 3 and macrophage colony-stimulating factor 1 correlate with disease activity in ankylosing spondylitis. Arthritis Rheum 2004, 51:691–9.

33. Pedersen SJ, Sørensen IJ, Lambert RGW, Hermann K-G a, Garnero P, Johansen JS, Madsen OR, Hansen A, Hansen MS, Thamsborg G, Andersen LS, Majgaard O, Loft AG, Erlendsson J, Asmussen KH, Jurik AG, Møller J, Hasselquist M, Mikkelsen D, Østergaard M: Radiographic progression is associated with resolution of systemic inflammation in patients with axial spondylarthritis treated with tumor necrosis factor α inhibitors. Arthritis Rheum 2011, 63:3789–800.

34. Maksymowych WP, Landewé R, Conner-Spady B, Dougados M, Mielants H, van der Tempel H, Poole a R, Wang N, van der Heijde D: Serum matrix metalloproteinase 3 is an independent predictor of structural damage progression in patients with ankylosing spondylitis. Arthritis Rheum 2007, 56:1846–53.

35. Diarra D, Stolina M, Polzer K, Zwerina J, Ominsky MS, Dwyer D, Korb A, Smolen J, Hoffmann M, Scheinecker C, van der Heide D, Landewe R, Lacey D, Richards WG, Schett G: Dickkopf-1 is a master regulator of joint remodeling. Nat Med 2007, 13:156–63.

36. Klingberg E, Nurkkala M, Carlsten H, Forsblad-d’Elia H: Biomarkers of bone metabolism in ankylosing spondylitis in relation to osteoproliferation and osteoporosis. J Rheumatol 2014, 41:1349–56.

37. Kwon S-R, Lim M-J, Suh C-H, Park S-G, Hong Y-S, Yoon B-Y, Kim H-A, Choi H-J, Park W: Dickkopf-1 level is lower in patients with ankylosing spondylitis than in healthy people and is not influenced by anti-tumor necrosis factor therapy. Rheumatol Int 2012, 32:2523–7.

38. Heiland GR, Appel H, Poddubnyy D, Zwerina J, Hueber A, Haibel H, Baraliakos X, Listing J, Rudwaleit M, Schett G, Sieper J: High level of functional dickkopf-1 predicts protection from syndesmophyte formation in patients with ankylosing spondylitis. Ann Rheum Dis 2012, 71:572–4.

39. Appel H, Ruiz-Heiland G, Listing J, Zwerina J, Herrmann M, Mueller R, Haibel H, Baraliakos X, Hempfing A, Rudwaleit M, Sieper J, Schett G: Altered skeletal expression of sclerostin and its link to radiographic progression in ankylosing spondylitis. Arthritis Rheum 2009, 60:3257–62.

40. Taylan A, Sari I, Akinci B, Bilge S, Kozaci D, Akar S, Colak A, Yalcin H, Gunay N, Akkoc N: Biomarkers and cytokines of bone turnover: extensive evaluation in a cohort of patients with ankylosing spondylitis. BMC Musculoskelet Disord 2012, 13:191.

41. De Andrade KR, de Castro GRW, Vicente G, da Rosa JS, Nader M, Pereira IA, Fröde TS: Evaluation of circulating

34 SERUM BIOMARKERS IN PSORIASIS AND IBD

levels of inflammatory and bone formation markers in axial spondyloarthritis. Int Immunopharmacol 2014, 21:481–6.

42. Korkosz M, Jerzy G, Leszczy P, Pawlak-bu K, Jeka S, Kucharska E: High disease activity in ankylosing spondylitis is associated with increased serum sclerostin level and decreased wingless protein-3a signaling but is not linked with greater structural damage. BMC Musculoskelet Disord 2013;14:99.doi:10.1186/1471-2474-14-99.

43. Chou C-L, Wu M-J, Yu C-L, Lu M-C, Hsieh S-C, Wu T-H, Chou C-T, Tsai C-Y: Anti-agalactosyl IgG antibody in ankylosing spondylitis and psoriatic arthritis. Clin Rheumatol 2010, 29:875–81.

44. Wright C, Sibani S, Trudgian D, Fischer R, Kessler B, Labaer J, Bowness P: Detection of Multiple Autoantibodies in Patients with Ankylosing Spondylitis Using Nucleic Acid Programmable Protein Arrays. Mol Cell Proteomics 2012:1–10.

45. Duftner C, Dejaco C, Klauser a, Falkenbach a, Lakomek HJ, Schirmer M: High positive predictive value of specific antibodies cross-reacting with a 28-kDa Drosophila antigen for diagnosis of ankylosing spondylitis. Rheumatology (Oxford) 2006, 45:38–42.

46. Baerlecken NT, Nothdorft S, Stummvoll GH, Sieper J, Rudwaleit M, Reuter S, Matthias T, Schmidt RE, Witte T: Autoantibodies against CD74 in spondyloarthritis. Ann Rheum Dis 2014, 73:1211–4.

47. Baraliakos X, Baerlecken N, Witte T, Heldmann F, Braun J: High prevalence of anti-CD74 antibodies specific for the HLA class II-associated invariant chain peptide (CLIP) in patients with axial spondyloarthritis. Ann Rheum Dis 2014, 73:1079–82.

48. 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, 65:12–20.

49. Fredriksson T, Pettersson U: Severe psoriasis--oral therapy with a new retinoid. Dermatologica 1978, 157:238–244.

50. Beygi S, Lajevardi V, Abedini R: C-reactive protein in psoriasis: a review of the literature. J Eur Acad Dermatol Venereol 2014, 28:700–11.

51. Coimbra S, Oliveira H, Reis F, Belo L, Rocha S, Quintanilha a., Figueiredo a., Teixeira F, Castro E, Rocha-Pereira P, Santos-Silva a.: C-reactive protein and leucocyte activation in psoriasis vulgaris according to severity and therapy. J Eur Acad Dermatology Venereol 2010, 24:789–796.

52. Nisa N, Qazi A: High-sensitivity C-reactive protein in psoriasis. Int J Dermatol 2012, 51:1393–1394.

53. Gisondi P, Malerbai M, Malara G, Guerra AP, Sala R, Radaelp A, Girolomoni G, Civili S: C-reactive protein and markers for thrombophilia in patients with chronic plaque psoriasis. Int J Immunopathol Pharmacol 2010, 23:1195–1202.

54. Strober B, Teller C, Yamauchi P, Miller JL, Hooper M, Yang Y-C, Dann F: Effects of etanercept on C-reactive protein levels in psoriasis and psoriatic arthritis. Br J

Dermatol 2008, 159:322–30.

55. Serwin AB, D M, Wasowicz W, Ph D, Chodynicka B, D M: Selenium supplementation , soluble tumor necrosis factor-alpha receptor type 1 , and C-reactive protein during psoriasis therapy with narrowband ultraviolet B. Nutrition 2006, 22:860–864.

56. Coimbra S, Oliveira H, Reis F, Belo L, Rocha S, Quintanilha a., Figueiredo a., Teixeira F, Castro E, Rocha-Pereira P, Santos-Silva a.: Circulating adipokine levels in Portuguese patients with psoriasis vulgaris according to body mass index, severity and therapy. J Eur Acad Dermatology Venereol 2010, 24:1386–1394.

57. Gisondi P, Lora V, Bonauguri C, Russo a, Lippi G, Girolomoni G: Serum chemerin is increased in patients with chronic plaque psoriasis and normalizes following treatment with infliximab. Br J Dermatol 2013, 168:749–55.

58. Rocha-Pereira P, Santos-Silva a, Rebelo I, Figueiredo a, Quintanilha a, Teixeira F: The inflammatory response in mild and in severe psoriasis. Br J Dermatol 2004, 150:917–28.

59. Coimbra S, Oliveira H, Reis F, Belo L, Rocha S, Quintanilha a, Figueiredo a, Teixeira F, Castro E, Rocha-Pereira P, Santos-Silva a: Interleukin (IL)-22, IL-17, IL-23, IL-8, vascular endothelial growth factor and tumour necrosis factor-α levels in patients with psoriasis before, during and after psoralen-ultraviolet A and narrowband ultraviolet B therapy. Br J Dermatol 2010, 163:1282–90.

60. Arican O, Aral M, Sasmaz S, Ciragil P: Serum levels of TNF-alpha, IFN-gamma, IL-6, IL-8, IL-12, IL-17, and IL-18 in patients with active psoriasis and correlation with disease severity. Mediators Inflamm 2005, 2005:273–9.

61. Flisiak I, Klepacki a, Chodynicka B: Plasma and scales levels of interleukin 18 in comparison with other possible clinical and laboratory biomarkers of psoriasis activity. Biomarkers 2006, 11:194–200.

62. Shimauchi T, Hirakawa S, Suzuki T, Yasuma A, Majima Y, Tatsuno K, Yagi H, Ito T, Tokura Y: Serum interleukin-22 and vascular endothelial growth factor serve as sensitive biomarkers but not as predictors of therapeutic response to biologics in patients with psoriasis. J Dermatol 2013, 40:805–12.

63. Borská L, Fiala Z, Krejsek J, Andrýs C, Vokurková D, Hamáková K, Ek JKČ, Ettler K: Selected Immunological Changes in Patients with sICAM-1 and IL-8. Physiol Res 2006, 55:699–706.

64. Elghandour TM, Youssef SES, Aly DG, Abd Elhameed MS, Abdel Moneim MM: Effect of Narrow Band Ultraviolet B Therapy versus Methotrexate on Serum Levels of Interleukin-17 and Interleukin-23 in Egyptian Patients with Severe Psoriasis. Dermatol Res Pract 2013, 2013:618269.

65. Caproni M, Antiga E, Melani L, Volpi W, Del Bianco E, Fabbri P: Serum levels of IL-17 and IL-22 are reduced by etanercept, but not by acitretin, in patients with psoriasis: a randomized-controlled trial. J Clin Immunol 2009, 29:210–4.

35

2

CHAPTER 2

66. Bonifati C, Trento E, Carducci M, Sacerdoti G, Mussi A, Fazio M, Ameglio F: Soluble E-selectin and soluble tumour necrosis factor receptor (60 kD) serum levels in patients with psoriasis. Dermatology 1995, 190:128–131.

67. Czech W, Schopf E, Kapp a.: Soluble E-selectin in sera of patients with atopic dermatitis and psoriasis-correlation with disease activity. Br J Dermatol 1996, 134:17–21.

68. Szepietowski J, Wa F, Bielicka E, Nockowski P, Noworolska A: Soluble E-selectin serum levels correlate with disease activity in psoriatic patients. Exp Dermatol 1999, 24:33–36.

69. Krasowska D, Chodorowska G, Kozioł M, Czelej D: Plasma levels of sICAM-1 in patients affected by psoriasis : no relation to disease severity. Med Sci Monit 2000, 6:353–355.

70. Gangemi S, Merendino RA, Guarneri F, Minciullo PL, Dilorenzo G, Pacor M, Cannavò SP: Serum levels of interleukin-18 and s-ICAM-1 in patients affected by psoriasis : preliminary considerations. Eur Acad Dermatology Venereol 2003:42–46.

71. Long JW, Tao J, Pi X, Wang YY: Effect of Narrow-band UVB Phototherapy on Soluble Cell Adhesion Molecules in Patients with Psoriasis Vulgaris. J Int Med reserach 2014, 38:1507–1512.

72. Madej a, Reich a, Orda a, Szepietowski JC: Vascular adhesion protein-1 (VAP-1) is overexpressed in psoriatic patients. J Eur Acad Dermatol Venereol 2007, 21:72–8.

73. Ataseven A, Kesli R: Novel inflammatory markers in psoriasis vulgaris: vaspin, vascular adhesion protein-1 (vap-1), and ykl-40. G Ital Dermatol Venereol 2014.

74. Nemati H, Khodarahmi R, Rahmani A, Ebrahimi A, Amani M, Eftekhari K: Serum lipid profile in psoriatic patients: correlation between vascular adhesion protein 1 and lipoprotein (a). Cell Biochem Funct 2013, 31(February 2012):36–40.

75. Schonthaler HB, Guinea-Viniegra J, Wculek SK, Ruppen I, Ximénez-Embún P, Guío-Carrión A, Navarro R, Hogg N, Ashman K, Wagner EF: S100A8-S100A9 protein complex mediates psoriasis by regulating the expression of complement factor C3. Immunity 2013, 39:1171–81.

76. Benoit S, Toksoy A, Ahlmann M, Schmidt M, Sunderkotter C, Foell D, Pasparakis M, Roth J, Goebeler M: Elevated serum levels of calcium-binding S 100 proteins A 8 and A 9 reflect disease activity and abnormal differentiation of keratinocytes in psoriasis. Clin Lab Investig 2006, 7:62–66.

77. Ataseven A, Kesli R, Kurtipek GS, Ozturk P: Assessment of lipocalin 2, clusterin, soluble tumor necrosis factor receptor-1, interleukin-6, homocysteine, and uric acid levels in patients with psoriasis. Dis Markers 2014, 2014:541709.

78. El-Hadidi H, Samir N, Shaker OG, Otb S: Estimation of tissue and serum lipocalin-2 in psoriasis vulgaris and its relation to metabolic syndrome. Arch Dermatol Res 2014, 306:239–45.

79. Kamata M, Tada Y, Tatsuta a, Kawashima T, Shibata S, Mitsui H, Asano Y, Sugaya M, Kadono T, Kanda N, Watanabe S, Sato S: Serum lipocalin-2 levels are increased in patients with psoriasis. Clin Exp Dermatol 2012, 37:296–9.

80. Romaní J, Caixàs A, Ceperuelo-Mallafré V, Carrascosa JM, Ribera M, Rigla M, Vendrell J, Luelmo J: Circulating levels of lipocalin-2 and retinol-binding protein-4 are increased in psoriatic patients and correlated with baseline PASI. Arch Dermatol Res 2013, 305:105–12.

81. Jansen P a M, Rodijk-Olthuis D, Hollox EJ, Kamsteeg M, Tjabringa GS, de Jongh GJ, van Vlijmen-Willems IMJJ, Bergboer JGM, van Rossum MM, de Jong EMGJ, den Heijer M, Evers AWM, Bergers M, Armour J a L, Zeeuwen PLJM, Schalkwijk J: Beta-defensin-2 protein is a serum biomarker for disease activity in psoriasis and reaches biologically relevant concentrations in lesional skin. PLoS One 2009, 4:e4725.

82. Gambichler T, Bechara FG, Scola N, Rotterdam S, Altmeyer P, Skrygan M: Serum levels of antimicrobial peptides and proteins do not correlate with psoriasis severity and are increased after treatment with fumaric acid esters. Arch Dermatol Res 2012, 304:471–4.

83. Johnston a, Arnadottir S, Gudjonsson JE, Aphale a, Sigmarsdottir a a, Gunnarsson SI, Steinsson JT, Elder JT, Valdimarsson H: Obesity in psoriasis: leptin and resistin as mediators of cutaneous inflammation. Br J Dermatol 2008, 159:342–50.

84. Baran a, Flisiak I, Jaroszewicz J, Swiderska M: Serum adiponectin and leptin levels in psoriatic patients according to topical treatment. J Dermatolog Treat 2014, 6634:1–5.

85. Karadag AS, Ertugrul DT, Kalkan G, Bilgili SG, Celik HT, Takci Z, Balahoroglu R, Calka O: The effect of acitretin treatment on insulin resistance, retinol-binding protein-4, leptin, and adiponectin in psoriasis vulgaris: a noncontrolled study. Dermatology 2013, 227:103–8.

86. Oh YJ, Lim HK, Choi JH, Lee JW, Kim NI: Serum leptin and adiponectin levels in Korean patients with psoriasis. J Korean Med Sci 2014, 29:729–34.

87. Zhu K-J, Zhang C, Li M, Zhu C-Y, Shi G, Fan Y-M: Leptin levels in patients with psoriasis: a meta-analysis. Clin Exp Dermatol 2013, 38:478–83.

88. Shibata S, Saeki H, Tada Y, Karakawa M, Komine M, Tamaki K: Serum high molecular weight adiponectin levels are decreased in psoriasis patients. J Dermatol Sci 2009, 55:62–3.

89. Ucak H, Demir B, Cicek D, Erden I, Aydin S, Dertlioglu SB, Arica M: Metabolic Changes and Serum Ghrelin Level in Patients with Psoriasis. Dermatol Res Pract 2014, 2014:1–6.

90. Ozdemir M, Yüksel M, Gökbel H, Okudan N, Mevlitoğlu I: Serum leptin, adiponectin, resistin and ghrelin levels in psoriatic patients treated with cyclosporin. J Dermatol 2012, 39:443–8.

91. Kawashima K, Torii K, Furuhashi T, Saito C, Nishio E, Nishida E, Shintani Y, Morita A: Phototherapy reduces serum resistin levels in psoriasis patients.

36 SERUM BIOMARKERS IN PSORIASIS AND IBD

Photodermatol Photoimmunol Photomed 2011, 27:152–5.

92. Shibata S, Tada Y, Hau C, Tatsuta a, Yamamoto M, Kamata M, Karakawa M, Asano Y, Mitsui H, Sugaya M, Kadono T, Saeki H, Kanda N, Sato S: Adiponectin as an anti-inflammatory factor in the pathogenesis of psoriasis: induction of elevated serum adiponectin levels following therapy. Br J Dermatol 2011, 164:667–70.

93. Bhushan M, McLaughlin B, Weiss JB, Griffiths CEM: Levels of endothelial cell stimulating angiogenesis factor and vascular endothelial growth factor are elevated in psoriasis. Br J Dermatol 1999, 141:1054–1060.

94. Meki A-RM a, Al-Shobaili H: Serum vascular endothelial growth factor, transforming growth factor β1, and nitric oxide levels in patients with psoriasis vulgaris: their correlation to disease severity. J Clin Lab Anal 2014, 28:496–501.

95. Flisiak I, Zaniewski P, Rogalska-Taranta M, Chodynicka B: Effect of psoriasis therapy on VEGF and its soluble receptors serum concentrations. J Eur Acad Dermatol Venereol 2012, 26:302–7.

96. Tekin NS, Ilter N, Sancak B, Ozden MG, Gurer MA: Nitric Oxide Levels in Patients with Psoriasis Treated with Methotrexate. Mediators Inflamm 2006, 2006:1–5.

97. Gokhale N, Belgaumkar V, Pandit D, Deshpande S, Damle D: A study of serum nitric oxide levels in psoriasis. Indian J Dermatol Venereol Leprol 2005, 71:175.

98. Zalewska A, Wyczółkowska J, Narbutt J, Sysa-Jędrzejowska A: Nitric oxide levels in plasma and fibroblast cultures of psoriasis vulgaris patients. J Dermatol Sci 2007, 48:237–240.

99. Serwin AB, Sokolowska M, Chodynicka B: Tumour necrosis factor a (TNF- alpha)-converting enzyme (TACE) and soluble TNF- a receptor type 1 in psoriasis patients treated with narrowband ultraviolet B. Photodermatol Photoimmunol Photomed 2007, 23:130–134.

100. Dekker-saeys BJ, Meuwissen SGM, Den EMVAN: IL . Prevalence of peripheral arthritis , sacroiliitis , and ankylosing spondylitis in patients suffering from inflammatory bowel disease. 1978:33–35.

101. Orchard TR, Wordsworth BP, Jewell DP: Peripheral arthropathies in inflammatory bowel disease: their articular distribution and natural history. Gut 1998, 42:387–391.

102. Palm O, Moum B, Ongre A, Gran JT, Palm Ø, Gran JANT: Patients with inflammatory bowel disease : a population study (the IBSEN study). 2002, 29.

103. Salvarani C: Clinical features and epidemiology of spondyloarthritides associated with inflammatory bowel disease. World J Gastroenterol 2009, 15:2449.

104. De Vos M, Cuvelier C, Mielants H, Veys E, Barbier F, A. E: Ileocolonoscopy in seronegative spondylarthropathy. Gastroenterology 1989:339–344.

105. Van Praet L, Van den Bosch FE, Jacques P, Carron P, Jans L, Colman R, Glorieus E, Peeters H, Mielants H, De Vos M, Cuvelier C, Elewaut D: Microscopic gut inflammation in axial spondyloarthritis: a multiparametric predictive model. Ann Rheum Dis 2013, 72:414–7.

106. Lees CW, Barrett JC, Parkes M, Satsangi J: New IBD genetics: common pathways with other diseases. Gut 2011, 60:1739–53.

107. Harvey D, Pointon JJ, Evans DM, Karaderi T, Farrar C, Appleton LH, Sturrock RD, Stone M a, Oppermann U, Brown M a, Wordsworth BP: Investigating the genetic association between ERAP1 and ankylosing spondylitis. Hum Mol Genet 2009, 18:4204–12.

108. Reveille JD, Sims A-M, Danoy P, Evans DM, Leo P, Pointon JJ, Jin R, Zhou X, Bradbury L a, Appleton LH, Davis JC, Diekman L, Doan T, Dowling A, Duan R, Duncan EL, Farrar C, Hadler J, Harvey D, Karaderi T, Mogg R, Pomeroy E, Pryce K, Taylor J, Savage L, Deloukas P, Kumanduri V, Peltonen L, Ring SM, Whittaker P, et al.: Genome-wide association study of ankylosing spondylitis identifies non-MHC susceptibility loci. Nat Genet 2010, 42:123–7.

109. Tremelling M, Cummings F, Fisher S a., Mansfield J, Gwilliam R, Keniry A, Nimmo ER, Drummond H, Onnie CM, Prescott NJ, Sanderson J, Bredin F, Berzuini C, Forbes A, Lewis CM, Cardon L, Deloukas P, Jewell D, Mathew CG, Parkes M, Satsangi J: IL23R Variation Determines Susceptibility But Not Disease Phenotype in Inflammatory Bowel Disease. Gastroenterology 2007, 132:1657–1664.

110. Bernstein CN, Fried M, Krabshuis JH, Cohen H, Eliakim R, Fedail S, Gearry R, Goh KL, Hamid S, Khan AG, LeMair a W, Malfertheiner, Ouyang Q, Rey JF, Sood A, Steinwurz F, Thomsen OO, Thomson A, Watermeyer G: World Gastroenterology Organization Practice Guidelines for the diagnosis and management of IBD in 2010. Inflamm Bowel Dis 2010, 16:112–24.

111. Best W, Becktel J, Singleton J, Jr. KF: Development of a Crohn’s disease activity index. National Cooperative Crohn’s Disease Study. Gastroenterology 1976, 70:439–444.

112. Walmsley RS, Ayres RC, Pounder RE, Allan RN: A simple clinical colitis activity index. Gut 1998, 43:29–32.

113. Schroeder K, Tremaine W, Ilstrup D: Coated Oral 5-Aminosalicylic Acid Therapy for Mildly to Moderately Active Ulcerative Colitis. N Engl J Med 1987, 317:1625–1629.

114. Poullis AP, Zar S, Sundaram KK, Moodie SJ, Risley P, Theodossi A, Mendall MA: A new , highly sensitive assay for C-reactive protein can aid the differentiation of in ¯ ammatory bowel disorders from constipation- and diarrhoea-predominant functional bowel disorders. Eur J Gastroenterol Hepatol 2002, 14:409–412.

115. Fagan E, Dyck R, Maton P, Hodgson H, Chadwick V, Petrie A, Pepys M: Serum levels of C-reactive protein in Crohn’s disease and ulcerative colitis. Eur J Clin Invest 1982, 12:351–359.

37

2

CHAPTER 2

116. Chamouard P, Richert Z, Meyer N, Rahmi G, Baumann R: Diagnostic value of C-reactive protein for predicting activity level of Crohn’s disease. Clin Gastroenterol Hepatol 2006, 4:882–7.

117. Jürgens M, Mahachie John JM, Cleynen I, Schnitzler F, Fidder H, van Moerkercke W, Ballet V, Noman M, Hoffman I, van Assche G, Rutgeerts PJ, van Steen K, Vermeire S: Levels of C-reactive protein are associated with response to infliximab therapy in patients with Crohn’s disease. Clin Gastroenterol Hepatol 2011, 9:421–7.e1.

118. Louis E, Vermeire S, Rutgeerts P, Vos M De, Gossum A Van, Pescatore P, Fiasse R, Pelckmans P, Reynaert H, Haens GD, Malaise M, Belaiche J, Louis E, Gastroente S De: A Positive Response to infliximab in crohn disease: Association with a higher systemic inflammation before treatment but not with - 308 TNF Gene Polymorphism. Scand J Clin Lab Invest 2002, 7:818–824.

119. Solem C a, Loftus E V, Tremaine WJ, Harmsen WS, Zinsmeister AR, Sandborn WJ: Correlation of C-Reactive Protein With Clinical, Endoscopic, Histologic, and Radiographic Activity in Inflammatory Bowel Disease. Inflamm Bowel Dis 2005, 11:707–712.

120. Wright J, Alp M, Young F, Tigler-Wybrandi N: Predictors of Acute Relapse of Crohn ’ s Disease A Laboratory and Clinical Study. Dig Dis Sci 1987, 32:164–170.

121. Gustot T, Lemmers a, Louis E, Nicaise C, Quertinmont E, Belaiche J, Roland S, Van Gossum a, Devière J, Franchimont D: Profile of soluble cytokine receptors in Crohn’s disease. Gut 2005, 54:488–95.

122. Koutroubakis IE, Petinaki E, Dimoulios P, Vardas E, Roussomoustakaki M, Maniatis AN, Kouroumalis E a: Increased serum levels of YKL-40 in patients with inflammatory bowel disease. Int J Colorectal Dis 2003, 18:254–9.

123. Vind I, Johansen J, Price P, Munkholm P: Serum YKL-40, a Potential New Marker of Disease Activity in Patients with Inflammatory Bowel Disease. Scand J Gastroenterol 2003, 38:599–605.

124. Erzin Y, Uzun H, Karatas A, Celik AF: Serum YKL-40 as a marker of disease activity and stricture formation in patients with Crohn’s disease. J Gastroenterol Hepatol 2008, 23(8 Pt 2):e357–62.

125. Herrlinger KR, Dittmann R, Weitz G, Wehkamp J, Ludwig D, Schwab M, Stange EF, Fellermann K: Serum Procalcitonin Differentiates Inflammatory Bowel Disease and Self-limited Colitis. Inflamm Bowel Dis 2004, 10:229–233.

126. Oruç N, Z ÖÖ, Lu NO, Lter T: Diagnostic value of serum procalcitonin in determining. Turkish J Gastroenterol 2009, 20:9–12.

127. Oussalah a, Laurent V, Bruot O, Guéant J-L, Régent D, Bigard M, Peyrin-Biroulet L: Additional benefit of procalcitonin to C-reactive protein to assess disease activity and severity in Crohn’s disease. Aliment Pharmacol Ther 2010, 32:1135–44.

128. Sachar D, Smith H, Chan S, Cohen L, Lichtiger S: Erythrocytic Sedimentation Rate as a measure of

clinical activity in iflammatory bowel disease. J Clin Gastroenterol 1986, 8:647–650.

129. Balding J, Livingstone WJ, Conroy J, Mynett-Johnson L, Weir DG, Mahmud N, Smith OP: Inflammatory bowel disease: the role of inflammatory cytokine gene polymorphisms. Mediators Inflamm 2004, 13:181–7.

130. Shanahan F: Crohn’s disease. Lancet 2002, 359:62–9.

131. Gross V, Andus T, Caesar I, Roth M: Evidence for continuous stimulation of interleukin-6 production in Crohn ’s disease. Gastroenterology 1992, 102:514–519.

132. Hyams JS, Fitzgerald JE, Treem WR, Wyzga N, Kreutzer DL: Relationship of functional and antigenic interleukin 6 to disease activity in inflammatory bowel disease. Gastroenterology 1993, 104:1285–1292.

133. Nancey S, Hamzaoui N, Moussata D, Graber I, Bienvenu J, Flourie B: Serum interleukin-6, soluble interleukin-6 receptor and Crohn’s disease activity. Dig Dis Sci 2008, 53:242–247.

134. Goke M, Hoffmann JC, Evers J, Kroger H, Manns MP: Elevated serum concentrations of soluble selectin and immunoglobulin type adhesion molecules in patients with inflammatory bowel disease. J Gastroenterol 1997, 32:480–486.

135. Jones SC, Banks RE, Haidar a, Gearing a J, Hemingway IK, Ibbotson SH, Dixon MF, Axon a T: Adhesion molecules in inflammatory bowel disease. Gut 1995, 36:724–730.

136. Magro F, Araujo F, Pereira P, Meireles E: Soluble Selectins , sICAM , sVCAM , and Angiogenic Proteins in Different Activity Groups of Patients with Inflammatory Bowel Disease. Dig Dis Sci 2004, 49(August):1265–1274.

137. Patel R, Abeed A, Dwomoa A, Keighley M: Circulating soluble adhesion molecules in inflammatory bowel disease. Eur J Gastroenterol Hepatol 1995.

138. Song W-B: Soluble intercellular adhesion molecule-1, D-lactate and diamine oxidase in patients with inflammatory bowel disease. World J Gastroenterol 2009, 15:3916.

139. Ates Y, Degertekin B, Erdil A, Yaman H, Dagalp K: Serum ghrelin levels in inflammatory bowel disease with relation to disease activity and nutritional status. Dig Dis Sci 2008, 53:2215–21.

140. Peracchi M, Bardella MT, Caprioli F, Massironi S, Conte D, Valenti L, Ronchi C, Beck-Peccoz P, Arosio M, Piodi L: Circulating ghrelin levels in patients with inflammatory bowel disease. Gut 2006, 55:432–3.

141. Sung EZH, Da Silva NF, Goodyear S, McTernan PG, Sanger GJ, Nwokolo CU: Increased plasma ghrelin following infliximab in Crohn’s disease. Aliment Pharmacol Ther 2009, 29:83–9.

142. Kader N, El-Din F, Khatab E, Elabd N: Does plasma resistin level have a role in predicting inflammatory bowel disease activity? Indian J Gastroenterol 2010, 29:126–127.

143. Konrad A, Lehrke M, Schachinger V, Seibold F, Stark R, Ochsenkuhn T, Parhofer KG, Goke B, Broedl UC:

38 SERUM BIOMARKERS IN PSORIASIS AND IBD

Resistin is an inflammatory marker of inflammatory bowel disease in humans. Eur J Gastroenterol Hepatol 2007, 19:1070–1074.

144. Morisaki T, Takeshima F, Fukuda H, Matsushima K, Akazawa Y, Yamaguchi N, Ohnita K, Isomoto H, Takeshita H, Sawai T, Fujita F, Nakao K: High serum vaspin concentrations in patients with ulcerative colitis. Dig Dis Sci 2014, 59:315–21.

145. Rodrigues VS, Milanski M, Fagundes JJ, Torsoni a S, Ayrizono MLS, Nunez CEC, Dias CB, Meirelles LR, Dalal S, Coy CSR, Velloso L a, Leal RF: Serum levels and mesenteric fat tissue expression of adiponectin and leptin in patients with Crohn’s disease. Clin Exp Immunol 2012, 170:358–64.

146. Karmiris K, Koutroubakis IE, Xidakis C, Polychronaki M, Voudouri T, Kouroumalis EA: Circulating levels of leptin, adiponectin, resistin, and ghrelin in inflammatory bowel disease. Inflamm Bowel Dis 2006, 12:100–105.

147. Weigert J, Obermeier F, Neumeier M, Wanninger J, Filarsky M, Bauer S, Aslanidis C, Rogler G, Ott C, Schaffler A, Scholmerich J, Buechler C: Circulating levels of chemerin and adiponectin are higher in ulcerative colitis and chemerin is elevated in Crohn’s disease. Inflamm Bowel Dis 2010, 16:630–637.

148. Franchimont D, Roland S, Gustot T, Quertinmont E, Toubouti Y, Gervy M-C, Deviere J, Van Gossum A: Impact of infliximab on serum leptin levels in patients with Crohn’s disease. J Clin Endocrinol Metab 2005, 90:3510–6.

149. Karmiris K, Koutroubakis IE, Xidakis C, Polychronaki M, Kouroumalis EA: The effect of infliximab on circulating levels of leptin, adiponectin and resistin in patients with inflammatory bowel disease. Eur J Gastroenterol Hepatol 2007, 19:789–794.

150. Manolakis AC, Kapsoritakis AN, Georgoulias P, Tzavara C, Valotassiou V, Kapsoritaki A, Potamianos SP: Moderate performance of serum S100A12, in distinguishing inflammatory bowel disease from irritable bowel syndrome. BMC Gastroenterol 2010, 10:118.

151. Brinar M, Cleynen I, Coopmans T, Van Assche G, Rutgeerts P, Vermeire S: Serum S100A12 as a new marker for inflammatory bowel disease and its relationship with disease activity. Gut 2010:1728–1730.

152. Schimid KW, Lugering N, Stoll R, Brinkb P, Winde G, Domschke W, Bocker W, Sorg C: Immunohistochemical Demonstration of the Calcium-Binding Proteins MRP8 and MRP14 and Their Heterodimer (27E 10 Antigen) in Crohn ’ s Disease. Hum Pathol 1995, 26:334–337.

153. Lugering N, Stoll R, Schmid K, Kucharzik T, Stein H, Burmeisterj G: The myeloic related protein MRP8/14 (27E10 antigen) - usefulness as a potential marker for disease activity in ulcerative colitis and putative biological function. Hum Pathol 1995, 14:659–664.

154. Meuwis M, Vernier-Massouille G, Grimaud JC, Bouhnik Y, Laharie D, Piver E, Seidel L, Colombel JF, Louis E: Serum calprotectin as a biomarker for Crohn’s disease. J Crohns Colitis 2013, 7:e678–83.

155. Leach ST, Yang Z, Messina I, Song C, Geczy CL, Cunningham AM, Day AS: Serum and mucosal S100 proteins, calprotectin (S100A8/S100A9) and S100A12, are elevated at diagnosis in children with inflammatory bowel disease. Scand J Gastroenterol 2007, 42:1321–31.

156. Malícková K, Kalousová M, Fucíková T, Bortlík M, Duricová D, Komárek V, Zima T, Janatková I, Lukás M: Anti-inflammatory effect of biological treatment in patients with inflammatory bowel diseases: calprotectin and IL-6 changes do not correspond to sRAGE changes. Scand J Clin Lab Invest 2010, 70:294–9.

157. Von Roon AC, Karamountzos L, Purkayastha S, Reese GE, Darzi AW, Teare JP, Paraskeva P, Tekkis PP: Diagnostic precision of fecal calprotectin for inflammatory bowel disease and colorectal malignancy. Am J Gastroenterol 2007, 102:803–13.

158. Van Rheenen PF, Van de Vijver E, Fidler V: Faecal calprotectin for screening of patients with suspected inflammatory bowel disease: diagnostic meta-analysis. BMJ 2010, 341:c3369.

159. Lin J-F, Chen J-M, Zuo J-H, Yu A, Xiao Z-J, Deng F-H, Nie B, Jiang B: Meta-analysis: fecal calprotectin for assessment of inflammatory bowel disease activity. Inflamm Bowel Dis 2014, 20:1407–15.

160. Wagner M: Fecal markers of inflammation used as surrogate markers for treatment outcome in relapsing inflammatory bowel disease. World J Gastroenterol 2008, 14:5584.

161. Sipponen T, Björkesten C-G a F, Färkkilä M, Nuutinen H, Savilahti E, Kolho K-L: Faecal calprotectin and lactoferrin are reliable surrogate markers of endoscopic response during Crohn’s disease treatment. Scand J Gastroenterol 2010, 45:325–31.

162. Kolho K-L, Sipponen T: The long-term outcome of anti-tumor necrosis factor-α therapy related to fecal calprotectin values during induction therapy in pediatric inflammatory bowel disease. Scand J Gastroenterol 2014, 49:434–41.

163. Hämäläinen A, Sipponen T, Kolho K-L: Infliximab in pediatric inflammatory bowel disease rapidly decreases fecal calprotectin levels. World J Gastroenterol 2011, 17:5166–71.

164. Ferreiro-iglesias R, Acosta MB, Santiago MO, Gonzalez L, Alonso C, Pen D, Estevez JB, Dominguez-mun JE: Fecal Calprotectin as Predictor of Relapse in Patients With Inflammatory Bowel Disease Under Maintenance. J Clin Gastroenterol 2015;00:000–000.

165. Wright EK, Kamm M a, De Cruz P, Hamilton AL, Ritchie KJ, Krejany EO, Leach S, Gorelik A, Liew D, Prideaux L, Lawrance IC, Andrews JM, Bampton P a, Jakobovits SL, Florin TH, Gibson PR, Debinski H, Macrae F a, Samuel D, Kronborg I, Radford-Smith G, Selby W, Johnston MJ, Woods R, Elliott PR, Bell SJ, Brown SJ, Connell WR, Day AS, Desmond P V, et al.: Measurement of fecal calprotectin improves monitoring and detection of recurrence of Crohn’s disease after surgery. Gastroenterology 2015, 148:938–947.e1.

39

2

CHAPTER 2

166. Janas RM, Ochocińska A, Snitko R, Dudka D, Kierkuś J, Teisseyre M, Najberg E: Neutrophil gelatinase-associated lipocalin in blood in children with inflammatory bowel disease. J Gastroenterol Hepatol 2014, 29:1883–9.

167. Oikonomou KA, Kapsoritakis AN, Theodoridou C, Karangelis D, Germenis A, Stefanidis I, Potamianos SP: Neutrophil gelatinase-associated lipocalin (NGAL) in inflammatory bowel disease: association with pathophysiology of inflammation, established markers, and disease activity. J Gastroenterol 2012, 47:519–530.

168. Yeşil A, Gönen C, Senateş E, Paker N, Gökden Y, Koçhan K, Erdem ED, Gündüz F: Relationship between neutrophil gelatinase-associated lipocalin (NGAL) levels and inflammatory bowel disease type and activity. Dig Dis Sci 2013, 58:2587–93.

169. Kim JM: Antimicrobial Proteins in Intestine and Inflammatory Bowel Diseases. Intest Res 2014, 9100:20–33.

170. Yamaguchi N, Isomoto H, Mukae H, Ishimoto H, Ohnita K, Shikuwa S, Mizuta Y, Nakazato M, Kohno S: Concentrations of alpha- and beta-defensins in plasma of patients with inflammatory bowel disease. Inflamm Res 2009, 58:192–7.

171. Zissis M, Afroudakis A, Galanopoulos G, Palermos I, Boura X, Sc B, Michopoulos S, Archimandritis A: B2 Microglobulin: Is It a Reliable Marker of Activity in Inflammatory Bowel Disease? 2001, 96.

172. Yılmaz B, Köklü S, Yüksel O, Arslan S: Serum beta 2-microglobulin as a biomarker in inflammatory bowel disease. World J Gastroenterol 2014, 20:10916–20.

173. Dotan I: New serologic markers for inflammatory bowel disease diagnosis. Dig Dis 2010, 28:418–23.

174. Homsak E, Micetić-Turk D, Bozic B: Autoantibodies pANCA, GAB and PAB in inflammatory bowel disease: prevalence, characteristics and diagnostic value. Wien Klin Wochenschr 2010, 122 Suppl :19–25.

175. Kuna AT: Review Serological markers of inflammatory bowel disease. Biochem Medica 2013, 23:28–42.

176. Bossuyt X: Serologic markers in inflammatory bowel disease. Clin Chem 2006, 52:171–81.

177. Prideaux L, De Cruz P, Ng SC, Kamm M a: Serological antibodies in inflammatory bowel disease: a systematic review. Inflamm Bowel Dis 2012, 18:1340–55.

178. Peeters M, Ph D, Joossens S, Sc M, Vermeire S, Vlietinck R, Bossuyt X, Rutgeerts P: Diagnostic Value of Anti- Saccharomyces cerevisiae and Antineutrophil Cytoplasmic Autoantibodies in Inflammatory Bowel Disease. Am J Gastroenterol 2001, 96:731–734.

179. Ruemmele FM, Targan SR, Levy G, Dubinsky M, Braun J, Seidman EG: Diagnostic accuracy of serological assays in pediatric inflammatory bowel disease. Gastroenterology 1998, 115:822–829.

180. Sandborn WJ, Loftus E V., Colombel JF, Fleming K a., Seibold F, Homburger H a., Sendid B, Chapman RW, Tremaine WJ, Kaul DK, Wallace J, Harmsen WS,

Zinsmeister AR, Targan SR: Evaluation of Serologic Disease Markers in a Population-Based Cohort of Patients with Ulcerative Colitis and Crohn’s Disease. Inflamm Bowel Dis 2001, 7:192–201.

181. Esters N, Vermeire S, Joossens S, Noman M, Louis E, Belaiche J, De Vos M, Van Gossum a, Pescatore P, Fiasse R, Pelckmans P, Reynaert H, Poulain D, Bossuyt X, Rutgeerts P: Serological markers for prediction of response to anti-tumor necrosis factor treatment in Crohn’s disease. Am J Gastroenterol 2002, 97:1458–62.

182. Smith BRK, Arnott IDR, Drummond HE, Nimmo ER: Disease location, anti-saccharomyces cerevisiae Antibody, and NOD2/CARD15 genotype influence the progression of disease behavior in crohn’s disease. Inflamm Bowel Dis 2004, 10:521–528.

183. Bonneau J, Dumestre-Perard C, Rinaudo-Gaujous M, Genin C, Sparrow M, Roblin X, Paul S: Systematic review: New serological markers (anti-glycan, anti-GP2, anti-GM-CSF Ab) in the prediction of IBD patient outcomes. Autoimmun Rev 2014, 14:231–245.

184. Karmiris K, Koutroubakis IE, Kouroumalis E a: Leptin, adiponectin, resistin, and ghrelin--implications for inflammatory bowel disease. Mol Nutr Food Res 2008, 52:855–66.

185. Ates Y, Degertekin B, Erdil A, Yaman H, Dagalp K: Serum ghrelin levels in inflammatory bowel disease with relation to disease activity and nutritional status. Dig Dis Sci 2008, 53:2215–2221.

186. Sari I, Kebapcilar L, Taylan A, Bilgir O, Kozaci DL, Yildiz Y, Yuksel A, Gunay N, Akkoc N: Fetuin-A and interleukin-18 levels in ankylosing spondylitis. Int J Rheum Dis 2010, 13:75–81.

187. Wendling D, Racadot E, Auge B, Toussirot E: Soluble intercelulular adhesion molecule 1 in spondyloarthritis. Clin Rheumatol 1998, 17:202–204.

188. Kemeny-Beke A, Gesztelyi R, Bodnar N, Zsuga J, Kerekes G, Zsuga M, Biri B, Keki S, Szodoray P, Betera A. Increased production of asymmetric dimethylarginine (ADMA) and atherosclerotic disease in ankylosing spondylitis. Joint bone spine 2011, 78:184–187.

189. Erre G, Sanna P, Zinellu A, Ponchietti A, Fenu P, Sotgia S, Carru C, Gannau A, Passiu G: Plasma asymmetric dimethylarginine (ADMA) levels and atherosclerotic disease in akylosing spondylitis: A cross-sectional study. Clin Rheumatol 2011, 30:21–27.

190. O’Shea F, Tsui F, Chiu B, Tsui H, Yazdanpanah M, Innman R: Retinol (vitamin A) and retinol binding protein levels are decreased in ankylosing spondylitis: clinical and genetic analysis. J Rheumatol 2007, 34:2457–2459.

191. Genre F, Lopez-Mejias R, Miranda-Filloy J, Ubilla B, Carnero-Lopez B, Gomez-Acebo I, Blanco R, Ochoa R, Rueda-Gotor J, Pina T, Gozalez-Juanatay C, Llorca J, Gonzalez-Gay M: Antitumour necrosis factor alpha treatment reduces retinol-binding protein 4 serum levels in non-dianetic ankylosing spondylitis patients. Ann Rheum Dis 2014, 73:941–943.

192. Ozgocmen S, Sogus S, Ardicoglu O, Fadillioglu E, I P, Akyol O: Serum nitric oxide, catalase, superoxide

40 SERUM BIOMARKERS IN PSORIASIS AND IBD

dismtase and malondialdehyde stats in patients with ankylsing spondylitis. Rheumatol Int 2004, 24:80–83.

193. Kozaci L, Sari I, Alacacioglu A, Akar S, Akkoc N: Evaluation of inflammation and oxidative stress in ankylosing spondylitis: a role for macrophage migration inhibitory factor. Mod Rheumatol 2010, 20:34–39.

194. Hoffman I, Dmetter P, Peeters M, De Vos M, Mielants H, Veys E, De Keyser F: Anti-saccharomyces cerevisiae IgA antibodies are raised in ankylosing spondylitis and undifferentiated spondyloarthropathy. Ann Rheum Dis 2003, 62:455–459.

195. Aydin S, Atugunduz P, Temel M, Bicakcigil M, Tasan D, Direskeneli H: Anti-saccharomyces cerevisiae antibodies (ASCA) in spondyloarthropathies: a reassessment. Rheumatology (Oxford) 2008, 47:142–144.

196. De Vries M, van der Horst-Bruinsma I, van Hoogstraten I, van Bodegraven A, von Blomberg BME, Ratnawati H, Dijkmans B: pANCA, ASCA, and OmpC antibodies in patients with ankylosing spondylitis without inflammatory bowel disease. J Rheumatol 2010, 37:2340–2344.

197. Appel H, Maier R, Wu P, Scheer R, Hempfing A, Kayser R, Thiel A, Radbruch A, Loddenkemper C, Sieper J: Analysis of IL-17(+) cells in facet joints of patients with spondyloarthritis suggests that the innate immune pathway might be of greater relevance than the Th17-mediated adaptive immune response. Arthritis Res Ther 2011, 13:R95.

198. Di Meglio P, Di Cesare A, Laggner U, Chu C-C, Napolitano L, Villanova F, Tosi I, Capon F, Trembath RC, Peris K, Nestle FO: The IL23R R381Q gene variant protects against immune-mediated diseases by impairing IL-23-induced Th17 effector response in humans. PLoS One 2011, 6:e17160.

199. Sarin R, Wu X, Abraham C: Inflammatory disease protective R381Q IL23 receptor polymorphism results in decreased primary CD4 + and CD8 + human T-cell functional responses. Proc Natl Acad Sci 2011, 108:9560–9565.

200. Raychaudhuri SK, Saxena A, Raychaudhuri SP: Role of IL-17 in the pathogenesis of psoriatic arthritis and axial spondyloarthritis. Clin Rheumatol 2015, 34:1019–23.

201. DeLay M, Turner M, Klenk E: HLA-B27 misfolding and the unfolded protein response augment interleukin-23 production and are associated with Th17 activation in transgenic rats. Arthritis Rheum 2009, 60:2633–2643.

202. Zhang L, Li Y, Li Y, Qi L, Liu X, Yuan C, Hu N, Ma D, Li Z, Yang Q, Li W, Li J: Increased frequencies of Th22 cells as well as Th17 cells in the peripheral blood of patients with ankylosing spondylitis and rheumatoid arthritis. PLoS One 2012, 7:e31000.

203. Kollnberger S, Bird L, Roddis M: HLA-B27 heavy chain homodimers are expressed in HLA-B27 transgenic rodent models of spondyloarthritis and are ligands for paired Ig-like receptors. J Immunol 2004, 173:1699–710.

204. Smith J, Turner M, DeLay M: Endoplasmic reticulum

stress and the unfolded protein response are linked to synergistic IFN-β induction via X-box binding protein. Eur J immmunology 2008, 38:1194–203.

205. Van Duivenvoorde LM, Dorris ML, Satumtira N, van Tok MN, Redlich K, Tak PP, Taurog JD, Baeten DL: Relationship between inflammation, bone destruction, and osteoproliferation in the HLA-B27/human β2 -microglobulin-transgenic rat model of spondylarthritis. Arthritis Rheum 2012, 64:3210–9.

206. Tran TM, Dorris ML, Satumtira N, Richardson J a, Hammer RE, Shang J, Taurog JD: Additional human beta2-microglobulin curbs HLA-B27 misfolding and promotes arthritis and spondylitis without colitis in male HLA-B27-transgenic rats. Arthritis Rheum 2006, 54:1317–27.

207. Hammer RE, Maika SD, Richardson JA, Tang J, Taurog JD: Spontaneous Inflammatory Disease in Transgenic Rats Expressing HLA-B27 and Human Pzm : An Animal Model of HLRB27-Associated Human Disorders. Cell 1990, 63:1099–1112.7