20
THE NEXT GENERATION IBD DIAGNOSTIC TEST The Synergistic Role of Serology, Genetics, and Inflammation in the Diagnosis of INFLAMMATORY BOWEL DISEASE

The Synergistic Role of Serology, Genetics, and ... NEXT GENERATION IBD DIAGNOSTIC TEST The Synergistic Role of Serology, Genetics, and Inflammation in the Diagnosis of INFLAMMATORY

Embed Size (px)

Citation preview

T H E N E X T G E N E R A T I O N I B D D I A G N O S T I C T E S T

The Synergistic Role of Serology, Genetics,and Inflammation in the Diagnosis of

INFLAMMATORY BOWEL DISEASE

INSIDE FRONT COVER(BLANK)

1

multiple imaging techniques are available to aid in the diagnosis of IBD,

many are associated with shortcomings from the standpoint of

performance. Computed tomographic enterography (CTE) is able to

detect clinically occult inflammatory and penetrating disease, but has

been reported to have less sensitivity than capsule endoscopy (CE) in

detecting ileal inflammation in patients with non-stricturing disease.

CTE sensitivity in detecting active colon inflammation has been estimated

at 74%. Magnetic resonance enterography (MRE) is associated with

suboptimal images in some patients. Like CTE, its ability to detect

inflammation is variable, with sensitivity ranging from 55% to 62%.

CE has the sensitivity to detect mucosal inflammation of the small bowel,

but its use can be complicated by capsule retention due to stricturing

disease and may lead to surgical intervention for a retained capsule.9,10

In patients with suspected or established CD, the diagnostic yield of CE

obtained from case reports, retrospective reviews, and prospective

evaluations ranges from 28% to 71%. Ileocolonoscopy is considered the

best test for detection of inflammation of the colon and has the added

benefit of allowing the collection of biopsy samples for pathologic

review. Biopsy results may not prove definitive, however, and the invasive

nature of the procedure remains a consideration, especially in the

pediatric age group. Ileocolonoscopy is associated with a small risk

of perforation (0.03% to 0.15%), as reported in a recent review of nine

large clinical series, each of which contained more than 30,000 cases.11

The need for an adjunctive and comprehensive testing tool is underscored

by the challenges related to diagnosing IBD and differentiating CD

from UC using a single modality, and with the knowledge that has

been gained regarding the known derangements present in IBD (eg,

inflammation, immune response to enteric bacteria, genetic factors

leading to IBD susceptibility).

Distinguishing Between CD and UC

Differentiating CD from UC is important for its implications in

selecting treatment and in the timing and type of surgery that may

ultimately be required. However, this differentiation remains a

challenge. A recent systematic evaluation by the Swiss IBD Study

Group in nearly 1600 patients with diagnoses of CD, UC, or

indeterminate colitis (IC) found that diagnostic delay was significantly

longer for CD than UC (median 9 vs 4 mo; P< .001).12 The intervals

between first symptoms to physician visit (P= .002), physician visit to

diagnosis (P < .001), and first symptoms to IBD diagnosis (P< .001)

were all significantly longer for patients with CD than for patients with

UC.12 On multivariate regression analysis, the presence of ileal

disease and age less than 40 years at diagnosis were found to be

independent risk factors for long diagnostic delay (>24 mo) in

patients with CD.12 In patients with UC, use of nonsteroidal anti-

inflammatory drugs was related to a prolonged diagnostic delay

(>12 mo) at a trend-level association (P= .093).12

Introduction

Inflammatory bowel disease (IBD) is a chronic and relapsing

idiopathic disorder of the gastrointestinal (GI) tract that is characterized

by mucosal inflammation and marked by recurrent diarrhea and

abdominal pain.1 While the pathogenesis of IBD is not completely

understood, current thinking is that it arises from complex interactions

involving the immune system, enteric commensal bacteria, and

genetic factors.2,3 IBD is divided into two main subtypes of IBD —

Crohn’s disease (CD) and ulcerative colitis (UC). While presenting

symptoms may be suggestive of one or the other entity, there can be

a large overlap of GI symptoms, associated physical findings, and

extraintestinal manifestations that are found in both UC and CD that

make it difficult to differentiate the two disorders,4 and thus there is a

medical need for adjunctive testing tools to aid clinicians with these

diagnostic challenges. This monograph outlines some of the

limitations of current IBD diagnostic testing modalities and describes

a newly available serologic, genetic, and inflammation biomarker

panel that may prove to be beneficial in this process.

Diagnostic Challenges in IBD

Establishing the IBD Diagnosis

At present, there is no “gold standard” test to definitively diagnose

IBD.4 Traditionally, the diagnosis of IBD is based on a combination of

data obtained from the patient history and physical examination in

association with laboratory, endoscopic, histologic, and radiographic

investigations. In adult and pediatric patients, presenting symptoms of IBD

are related to both disease location and extent of intestinal inflammation.5

However, even when symptoms suggest a diagnosis of IBD, they are

often too nonspecific to result in a definitive diagnosis, such as in the

case of symptom overlap with functional bowel disorders6 or infectious

colitides.4 This overlap frequently delays an accurate diagnosis of IBD.6

Despite the lack of a pathognomonic test for IBD, laboratory values

provide useful information in the diagnostic process. Hematologic

parameters (increased leukocyte and thrombocyte counts), elevated

levels of acute phase reactants (C-reactive protein [CRP] and

erythrocyte sedimentation rate), and the presence of fecal markers

(leukocytes, lactoferrin, calprotectin) may be associated with active

intestinal inflammation,7,8 although they do not identify an exact

underlying cause. As a result of the limited negative predictive value

of laboratory values, the differential diagnosis requires additional

testing to rule out alternative disorders including other inflammatory

bowel diseases such as those related to infectious agents, medications,

radiation, or ischemia; other intestinal disorders (eg, celiac disease,

microscopic colitis); or irritable bowel syndrome (IBS).7

Establishing the diagnosis of IBD generally relies on the use of

invasive testing such as imaging studies and endoscopy. Although

2

Differentiating CD from UC can be difficult in patients whose disease

manifestations are exclusively colonic in nature and who lack typical

endoscopic or histologic findings, such as in patients with extensive

acute, severe colitis.13 While the main pathologic criterion for the diagnosis

of CD is the presence of granulomas, these may not be universally

evident in tissue samples obtained either at surgery or at biopsy. A

retrospective chart review including 102 patients with CD by Wolfson

and colleagues found granulomas in only 45% to 58% of surgically

resected specimens examined.14 An even smaller percentage (as low

as 2% to over 20%) is reported to be found in biopsy samples.15

Evaluation of Populations at Risk

Genetic susceptibility has been implicated in the pathogenesis of

IBD.2 Twin studies carried out in Sweden and Denmark revealed UC

concordance rates of 6.3% to 18.2% in monozygotic twins and 3.9%

to 4.5% in dizygotic twins.16,17 In these same studies, concordance in

CD was high for monozygotic twins (58.3%) but not for dizygotic

twins (0%).16,17 Furthermore, multiple investigations have shown that

first-degree relatives of patients with IBD are at an increased risk of

developing IBD. In a 2004 review of such studies, Russell and Satsangi

reported that the risk of developing CD was highest in siblings of CD

probands, where the relative risk was 30 to 42, whereas the overall

relative risk for CD in any first-degree relative was 10 to 35.18 The

relative risk of developing UC in first-degree relatives of UC probands

ranged from 10 to 15.18

Results of family and twin studies suggesting a contribution of

genetic factors to IBD prompted the search for susceptibility genes in

the 1990s. Initial efforts in genetic linkage studies and candidate gene

approaches in IBD were slow and gave rise to “susceptibility gene”

replication studies.

In 2000, efforts to sequence the human genome were followed by the

discovery of more than 10 million single nucleotide polymorphisms

(SNPs), which are DNA sequence variations that occur when a single

nucleotide (A, T, C, or G) in the genome sequence is altered.19 Many

SNPs have no effect on cell function, but scientists believe others could

predispose people to disease or influence their response to a drug.19

Together, these scientific advances made possible a new type of

research effort—the genome-wide association study (GWAS). In a

GWAS, the distribution of SNPs is determined in large populations

with and without a specific disease. By determining which SNPs

co-occur with disease symptoms, researchers can make a statistical

estimate of the level of risk associated with each SNP. This cutting-

edge research has resulted in the discovery of many susceptibility

genes in IBD. Furthermore, genes that confer susceptibility to one or

the other constituent diseases—either CD or UC—have also been

elucidated, providing a useful tool for diagnostic differentiation.

Evolution of IBD Testing: PROMETHEUS Laboratories, Inc.

Biomarkers that differentiate IBD from functional and other bowel

disorders, and that can distinguish UC from CD, have the potential to

improve the diagnostic process and minimize complications resulting

from invasive diagnostic procedures. These biomarkers may be

especially advantageous in a pediatric population.

Initially, two serologic markers—ASCA (anti-Saccharomyces

cerevisiae antibodies) and pANCA (perinuclear antineutrophil

cytoplasmic antibodies) — showed utility in diagnosing IBD and

differentiating between UC and CD.20 ASCA is more prevalent in CD than

in UC and healthy controls,21 and pANCA has more specificity for UC.20

However, some overlap was noted in that about one fourth of CD patients

will express pANCA,22 and ASCA sensitivity is suboptimal for a definitive

diagnosis of CD despite its high specificity. Moreover, pANCA is not

highly sensitive for UC because of its overlap in expression in UC-like

CD.20 The low sensitivity of ASCA or pANCA in IBD limits their clinical

utility in that a negative test result can rule out neither CD nor UC.23

Whereas positivity for ASCA and/or pANCA may help assign IC patients

a definitive diagnoses of CD or UC, a large percentage of IC patients

(85.1%) have been found to be seronegative for both markers.24

Efforts to improve the sensitivity of serologic testing in IBD have

led to the discovery and validation of new biomarkers and the

evolution of diagnostic test panels (Figure 1).

The IBD First Step (PROMETHEUS Laboratories, Inc., San Diego, CA)

improved the sensitivity of assays for ASCA and pANCA and added a

new biomarker, anti-OmpC (antibody to outer membrane porin C), to

increase sensitivity for CD.20 The next generation of this test, IBD

Serology 7, was augmented with the recently discovered biomarker

anti-CBir1 and incorporated the use of a Smart Diagnostic Algorithm

to interpret the overall (versus individual) biomarker interaction

results, thus providing a more accurate assessment than previous

serologic assays.20

In efforts to continue to improve the performance of testing for

IBD, as well as the ability to better differentiate between CD and

UC, PROMETHEUS Laboratories has now developed the novel

and innovative PROMETHEUS IBD sgi Diagnostic test. Designed as

an adjunctive test for the workup of suspected IBD and for the

differentiation of CD and UC in known IBD, the PROMETHEUS IBD sgi

Diagnostic test contains three major classes of biomarkers —serologic,

genetic, and inflammatory — that correspond to the complex

multidimensional nature of IBD (Figure 2). Genetic markers chosen to

be included in the PROMETHEUS IBD sgi Diagnostic test have relevance

to various physiologic aspects known to be compromised or impaired in

IBD, thus leading to disease susceptibility. Specifically these processes

include defective bacterial handling/autophagy, dysregulated signaling

pathways, and impaired epithelial barrier function.

3

Figure 1. Evolution of Diagnostic Testing in IBD

Earlybiomarkertesting

ASCA pANCA

ASCA pANCAAnti-OmpC

ASCA-IgAASCA-IgGAnti-CBir1-IgAANCA-IgGAnti-OmpC-IgApANCADNase-sensitive pANCA

ASCA-IgAASCA-IgGANCA-IgGpANCADNase-sensitive pANCAAnti-CBir1 Anti-OmpC Anti-A4-Fla2Anti-Fla-X

IBD sgi Diagnostic

IBD Serology 7

IBD First Step

STAT3ECM1NKX2.3ATG16L1VEGFICAMVCAMCRPSAA

Figure 2. IBD occurs under the influence of commensal enteric microflora and mucosal immunity (s) as well as environmental factors in the

intestines of genetically susceptible individuals (g) in the presence of inflammatory markers (i).

ANCA, antineutrophil cytoplasmic antibodies; anti-OmpC, antibodies to outer membrane porin C; ASCA, anti-Saccharomyces cerevisiae antibodies;

ATG16L1, autophagy related 16-like 1; CRP, C-reactive protein; DNase, deoxyribonuclease; ECM1, extracellular matrix protein 1; ICAM, intercellular

adhesion molecule; IgA, immunoglobulin A; IgG, immunoglobulin G; pANCA, perinuclear antineutrophil cytoplasmic antibodies; SAA, serum amyloid A;

STAT, signal transducers and activators of transduction; VCAM, vascular cell adhesion molecule; VEGF, vascular endothelial growth factor.

4

The text that follows describes the specific serologic, genetic, and

inflammatory markers that make up the PROMETHEUS IBD sgi

Diagnostic test.

Serologic Markers

Serologic markers are important in CD because their expression

represents the host response to translocation of intestinal pathogens

into the bloodstream as a result of breakdown of the gut mucosal

barrier. Investigations to uncover the underlying cause of the abnormal

intestinal inflammatory reaction that characterizes IBD have led to the

discovery of antibodies that are present specifically in the blood of

patients with CD or UC.25 Initially, research focused on ASCA, which

was associated with CD, and pANCA, which was associated with UC.

Given the heterogeneity of IBD, it soon became clear that these

biomarkers would not lead to a diagnosis of IBD in all patients who

have the disorder, as about 30% are negative for both ASCA and

pANCA immune responses.26 Additionally, it was noted that a subset of

CD patients have seroreactivity to pANCA and phenotypically have

“UC-like CD.” Continued research expanded the number of serologic

markers that are associated with IBD and/or have the capacity to better

differentiate CD and UC. The resulting set of serologic markers that now

compose the PROMETHEUS IBD sgi Diagnostic test are detailed below.

Yeasts

ASCA-IgA and -IgG

ASCA (anti-Saccharomyces cerevisiae antibodies) are antibodies

directed against mannose sequences from phosphopeptidomannan

comprising the cell wall of the baker’s and brewer’s yeast Saccharomyces

cerevisiae.21 Candida albicans has also been shown to express ASCA

epitopes on mannoproteins similar to that found in S cerevisiae.27

ASCA has been shown to correlate with C albicans colonization in

healthy relatives of patients with CD but not in CD patients themselves.28

The prevalence of ASCA positivity is highest in CD patients, ranging

from 35% to 76%, but also occurs in 5% to 15% of patients with UC

and in up to 5% of healthy controls.29,30

Bacterial Proteins

Anti-OmpC

OmpC (outer membrane porin C) is a bacterial antigen originally

isolated from Escherichia coli.31 This antigen results in

immunoglobulin G and A (IgG and IgA) antibody responses (anti-

OmpC)30 in 37% to 55% of adults with CD.32-34 In a cohort of

children with CD, 17% were found to have a seropositive response

to anti-OmpC.35 In another evaluation of antibody responses in

children and young adults (age ≤24.0 yr) with CD or UC, anti-OmpC

positivity was found in 25% of patients with CD, 11% of patients with

UC, and 5% of healthy controls.36 While anti-OmpC as a stand-alone

marker lacks sensitivity in CD and UC, the addition of this marker to

a panel that includes ANCA, ASCA-IgG, and ASCA-IgA improves

sensitivity for IBD. Indeed, a four-marker diagnostic panel including

anti-OmpC identified CD in 65% of children and UC in 74% of children,

with a 94% specificity.36

Anti-CBir1

The human intestinal tract is colonized by a vast assortment of

commensal microbial species, while also being occasionally exposed to

bacteria that are potentially pathogenic. Subsets of both commensal and

pathogenic bacteria are motile as a result of the expression of one or more

flagella. The primary structural component of bacterial flagella is the

35-50 kDa protein flagellin. The region of the flagellin molecule involved

in its polymerization to flagella is highly conserved among various

bacteria. Flagellin has been found to be a major target of both innate37

and adaptive immune responses38,39 that are associated with IBD.

In 2004, Lodes and colleagues used serologic expression cloning

to search for bacterial antigens relevant to IBD and identified flagellins

as among the immunodominant antigens of the microbiota.38 The specific

flagellin expression clone, CBir1, was subsequently noted to induce a

strong antibody response (anti-CBir1) in four genetically distinct

mouse models of IBD.38 In humans, these researchers noted significantly

higher levels of anti-CBir1 in patients with CD than in patients with UC

and controls, suggesting that this response might be useful in the

diagnosis of IBD and in the differentiation of CD and UC.38 Further

research by Targan and colleagues found that anti-CBir1 was expressed

in 50% to 55% of patients with CD.40 Controlling for the previously

described microbial antigens ASCA (-IgG and -IgA), anti-OmpC, and

anti-I2 (antibody to the I2 bacterial sequence found in Pseudomonas

fluorescens), these researchers found that anti-CBir1 was independently

associated with CD (P<.001) and its expression level was unrelated to

any one of the other four individual antimicrobial antibodies associated

with CD; as a result, anti-CBir1 reactivity defined another subpopulation

of CD patients that was pathophysiologically distinct.40 Anti-CBir1

expression was also found in approximately 50% of CD patients who

did not express ASCA; 40% to 44% of patients with CD whose sole

antibody response was to pANCA were found to express anti-CBir1

compared with only 4% of patients with UC who were otherwise solely

positive for pANCA.40 Furthermore, anti-CBir1 expression was found

in 40% of patients who lacked a serologic response to any of the

other antigens studied.40 More recently, Markowitz and colleagues

investigated immune responses in 705 children, age 0 to 15 years, with

CD. This group found that 52% were seronegative for pANCA, ASCA-

IgA and -IgG, and anti-OmpC.35 This percentage of seronegative

patients fell to 27% when assessment of anti-CBir1 was added

(P < .0001), thus identifying a significant subset of children with CD

who were otherwise seronegative.35

5

(ie, DNase-sensitive pANCA) appears to be a dominant characteristic

of UC-specific pANCA, distinguishing the disorder from primary

sclerosing cholangitis and type 1 autoimmune hepatitis.45

Genetic Markers

GWAS methodology has revolutionized research into IBD.46 The

GWAS approach is very effective in the detection of relatively common

gene variants and forms the basis for fine-mapping of identified genes

and research into their function. This has increased the knowledge of

the various signaling pathways involved in the disorder(s) and also

has implications for the development of future treatments. Genes that

have been definitively associated with IBD, CD, or UC are listed in

Figure 3 and the list is expanding rapidly.

ATG16L1

Defective bacterial handling has become one of the most consistent

focuses in the study of CD pathogenesis.46 One gene that is related to

the process of autophagy, ATG16L1 (autophagy related 16-like 1 gene),

has been associated with CD, autophagy has many physiologic roles

in health and disease, including acting as a “housekeeping” mechanism

by which cells digest parts of their own cytoplasm for removal or

providing an innate immune mechanism by which the host can

combat intracellular bacteria.47

ATG16L1 was first identified by Hampe and colleagues as a

susceptibility gene for CD in a GWAS of 19,779 non-synonymous single

nucleotide polymorphisms (SNPs) present in 735 patients with CD and

in 368 controls.48 Non-synonymous SNPs are SNPs that have different

alleles that encode different amino acids.49 Analysis showed that the

marker rs2241880 encoded a threonine-to-alanine substitution at amino

acid position 300 (T300A), which was correlated with the incidence of

CD in one British and two German studies of CD.48 No correlation

between the rs2241880 marker and UC was noted. Cummings and

colleagues50 attempted to replicate the findings of Hampe and

colleagues48 in a cohort of 648 CD cases, 677 UC cases, and 1190

controls that were well characterized. This group found that carriage

of the G risk allele of rs2241880 was strongly associated with CD

(P=2.33 × 10-7; odds ratio [OR] 1.45 [1.25–1.67]), thus reproducing

the Hampe nsSNP GWAS findings. A recent meta-analysis of 25 studies

(published up to June 1, 2009)51 evaluating the association of ATG16L1

and IBD found a positive association between the T300A polymorphism

and susceptibility to CD (OR 1.32; 95% confidence interval [CI]

1.26–1.39; P< .00001). ATG16L1 was also associated with the risk of

childhood-onset CD but not childhood-onset UC.51

The same polymorphism was recently evaluated in a Spanish cohort

comprising 557 CD and 425 UC patients compared with 672 ethnically

matched controls52 and was found to be strongly associated with

susceptibility to CD (OR 1.62) but showed no association with UC.

Anti-Fla-X and Anti-A4-Fla2

In their research, Lodes and colleagues cloned a second reactive

flagellin (Fla-X) that was highly homologous to CBir1 (83.5%

similarity at the NH2 conserved domain) and, similar to the findings

with CBir1, strong immune reactivity directed toward the NH2 terminal

end of Fla-X was noted in both a colitic mouse model and in patients

with CD.38 The level of response to Fla-X observed in sera from

patients with CD was significantly higher than that found in sera from

normal controls and patients with UC. Given that more than 50% of

the UC sera tested came from patients with a Truelove and Witts

severity index indicative of moderate to active disease (ie, >7), these

antiflagellin responses have potential not only in the diagnosis of IBD

but also in the discrimination of UC and CD.38

Since prior phylogenic analyses have linked the Clostridium

phylogenetic cluster XIVa to the origin of flagellins.41 Duck and colleagues

investigated this cluster and characterized a number of flagellated

bacteria from it.42 These investigators found that the A4 bacterial strain

expresses a flagellin, A4-Fla2, which has a very similar amino acid

sequence to the flagellin Fla-X43 to which patients with CD have

demonstrated seropositivity.38 Subsequently, Schoepfer and colleagues44

assessed seroreactivity to A4-Fla2 and Fla-X in a population of well-

characterized CD (n=252) or UC (n=53) patients and in healthy controls

(n=43). Seropositivity to A4-Fla2 and Fla-X was found in 59% and 57%

of patients with CD, respectively, 6% each in patients with UC, and 0%

and 2% of healthy controls, respectively.44 The serologic response

generated against either flagellin in patients with CD was most frequently

IgG, being found in 94% of those positive for anti-A4-Fla2 and in 93%

of those positive for anti-Fla-X. An IgA response was found in < 20% of

CD patients for either flagellin.44 No correlation was found between

disease duration and serologic response to A4-Fla2 or Fla-X. This

research has underscored the dominance of flagellin as an antigen in CD.

Given the slightly higher rate of seroreactivity to Fla-X and A4-Fla2 than

to CBir1 noted in CD patients, these two flagellins may enhance the

ability to diagnose CD over and above assessment of anti-CBir1.

Autoantibodies

ANCA, pANCA, and DNase-sensitive pANCApANCA (perinuclear antineutrophil cytoplasmic antibodies) are

autoantibodies directed against a component of neutrophil granules.30

Distinct subgroups of ANCAs have been noted with indirect

immunofluorescence based on their individual staining pattern.29 The

use of immunofluorescence has revealed a perinuclear pattern of

staining (pANCA) in association with many different diseases including

vasculitides and collagenous and eosinophilic colitides.23 The pANCA

staining pattern has also been found in 30% to 83% of patients with UC,

6% to 20% of patients with CD, and up to 2.5% of healthy controls.29,30

Loss of this antigenic response after DNase digestion of neutrophils

6

STAT3

The STAT (signal transducers and activators of transcription)-

Janus kinase pathway controls signal transduction between cell surface

receptors and the nucleus. STAT3, an inducible DNA binding protein that

binds to the interleukin (IL)-6 responsive element within the promoters

of hepatic acute phase protein genes,58 is activated by a wide variety of

cytokines and growth factors.59 Upon activation, STAT3 is phosphorylated,

resulting in dimerization; the dimer migrates to the nucleus where it

induces the expression of genes that have roles in many biologic

functions including cell growth, antiapoptosis and proapoptosis, cell

motility, negative feedback (suppression of cytokine production),

regulatory cytokine production, and antibacterial activity.59-62

It is well established that STAT3 is necessary for the signaling

of proinflammatory cytokines such as IL-6, which is involved in

the pathogenesis of both multiple sclerosis and IBD.63 STAT3 is

also important for multiple aspects of the biology of Th17 cells,

which, in turn, are critically important as mediators of a number of

human inflammatory diseases.64 In mouse models, STAT3 plays

distinct roles in both innate and acquired immunity. STAT3-mediated

activation of acquired immune response plays a pathogenic role in

NKX2.3

Signaling pathways that are critical to normal mammalian gut

development are dysregulated in IBD. NKX2.3 (NK2 transcription

factor-related, locus 3) is a member of the NKX family of

homeodomain-containing transcription factors implicated in cell

type specification and maintenance of differentiation in several

types of tissue.46 NKX2.3 is expressed in small blood vessel

endothelial cells and intestinal lamina propia mesenchymal cells.46

Evidence from mouse studies suggests that abnormal expression

of NKX2.3 alters the migration of antigen-responsive lymphocytes

in the gut and influences the inflammatory response in the

intestines.53 Susceptibility for CD was demonstrated with the

rs10883365 variant of NKX2.3 in multiple GWAS studies in which

ORs between 1.3 and 1.5 were observed.53-55 Additionally, in the first

non-synonymous SNP scan in UC, Fisher and colleagues found an

association between the rs10883365 NKX2.3 variant and UC

(P = 3.3×104).56 Meggyesi and colleagues later found this association

in a group of Czech and Hungarian IBD patients (increased risk for

UC, P= .003 after Bonferroni correction; OR 1.36; 95% CI 1.13–

1.63),57 demonstrating that NKX2.3 represents an IBD locus.

Figure 3. Genes Associated with Inflammatory Bowel Disease, Crohn’s Disease, and Ulcerative Colitis

(Adapted with permission from Thompson and Lees .68)

UC

IBD

CD

ECM1IL10IFN-gammaIL22FCGR2AIL2/IL21IL17REL/PIM3CAPN10/GPR35

LAMB1/SCL26A3OTUD3/PLA2G2EARPC2HNF4-alpha CDH1IL2613q121p36

CARD9CEP72/TPPP13q13SMURF1/KPNA7IFN-gamma/IL26/IL22

ATG16L1NOD2C11orf30ZNF365CCR61q246q217p12

13q14ICOSLGGPX4GCKRSLC22A23NLRP3PTPN22LRRK2

1q326q258q2418q11ITLN1ORMDL3CCL2/CCL7/CCL11/CCL8

STAT3NKX2.3IL23RIL12/p40JAK23p21/MST1REL/PUS10ZMIZ1MTMR3

IRGMORMDL321q21MHC*IL18RAPLYRM4CDKAL1TNFRSF6B (DCR3)PSMG1

CCNYIL27IBD5PUS10TNFSF15 (TL1A)19q13HORMAD2/MTMR3PTGER4

*Colonic disease

7

gut tissues causing gut inflammation and injury.74,75 Inflammatory

characteristics shared between patients with IBD and those with

other chronic immune disturbances include immune activation,

the infiltration of leukocytes into tissues, and an increased

vascular density.75

Intercellular Adhesion Molecule 1 and

Vascular Cell Adhesion Molecule 1

Dense inflammatory infiltrates, including monocytes,

lymphocytes, and neutrophils, characterize IBD and are seen in

distinct distributions in both CD and UC.76 Infiltration of these cells

is stimulated both by various cytokines and by the interactions

that occur between adhesion molecules expressed on inflammatory

cells in the circulation and their integrin receptors on localized

target cells.77

Intercellular adhesion molecule 1 (ICAM-1) and vascular cell

adhesion molecule 1 (VCAM-1) are cytokine-inducible cell surface

glycoproteins that belong to the immunoglobulin supergene

family.78 Soluble ICAM and VCAM are also detected in serum and

are believed to arise from proteolytic cleavage of the cell surface

molecules. ICAM-1 is expressed at low levels on endothelial,

epithelial, and other cells as well as on lymphocytes and

monocytes.76 The transcription of ICAM-1 is upregulated by

proinflammatory cytokines.79,80 In inflamed tissues, increased

expression of ICAM-1 on endothelial cells promotes the

recruitment of inflammatory cells expressing its ligands (any

substance that binds specifically and reversibly to another

chemical entity to form a larger complex), leukocyte function

antigen-1, or macrophage activation complex-1, thus propagating

the inflammatory process. In the rat, interferon-γ– induced

expression of ICAM-1 has been noted to be higher in deep

mucosa/submucosa and muscle layer microcirculations than in

the superficial mucosa—layers of the gut involved in the

characteristic pathology of CD—where it may contribute to

leukocyte-endothelial interactions.81

The expression of VCAM-1 is found on activated endothelial

cells, macrophages, dendritic cells, and fibroblasts. Ligands for

VCAM-1 include the integrins α4/β4 (very late antigen 4) and

α4/β7, which are present on monocytes, lymphocytes, and

eosinophils. Upon binding to its ligands, VCAM-1 modulates

leukocyte adhesion to endothelial cells and migration to sites of

inflammation.82,83

Jones and colleagues investigated circulating concentrations

of ICAM-1 and VCAM-1 in 43 patients with IBD (22 with CD and

21 with UC) compared with healthy controls (n=90).77 Circulating

colitis by enhancing survival of pathogenic T cells and by

inducing tumor necrosis factor alpha (TNF-α). In contrast,

STAT3-mediated activation of innate response contributes to the

suppression of colitis by enhancing the mucosal repair and

induction of mucin production.65

Using GWAS, Barrett and colleagues found that the rs744166

variant of STAT3 was associated with CD susceptibility.66 This

association was confirmed in a Spanish cohort of patients reported by

Cenit and colleagues.67 This group analyzed polymorphisms in the

STAT3 region (rs3809758/rs744166/rs1026916/rs12948909). The

haplotype conformed by the risk alleles of each polymorphism was

significantly associated with both CD (P= .005; OR 1.25; 95% CI

1.06–1.46) and UC (P= .002; OR 1.19; 95% CI 1.02–1.38).67

ECM1

One of the most intriguing concepts that has emerged regarding

the genetics of UC has been its association with epithelial barrier

genes.68 Intercellular junctions between intestinal epithelial cells play

a crucial gatekeeping role in the gut by allowing the extraction of

needed nutrients while keeping pathogens out of the systemic

circulation. These tight junctions determine overall intestinal

permeability. In IBD, epithelial permeability is enhanced and is a

predictor of clinical relapse.69,70

In the first non-synonymous SNP scan for UC, Fisher and

colleagues were able to identify a previously unknown

susceptibility locus, ECM1, in UC.56 ECM1 is an epithelial barrier

gene that encodes the glycoprotein ECM1 (extracellular matrix

protein 1), which is expressed in the small and large intestines

where it interacts with the basement membrane and inhibits

matrix metalloproteinase 9. ECM1 also strongly activates nuclear

factor-κB (NF-κB) signaling, which is a key component in a variety

of regulatory pathways.71 Expression of ECM1 is upregulated in

esophageal squamous cell carcinoma and colorectal cancer.72

While an association with the ECM1 variant has been found for UC,

it was not observed when formally tested in an adequate sample of

CD subjects, implying that ECM1 may provide a specific

susceptibility association for UC alone.73

Inflammatory Markers

The presence of intestinal inflammation is a primary criterion

for making the diagnosis of IBD. Under normal conditions, the

intestinal mucosa is in a state of “controlled” inflammation that is

regulated by a delicate balance between proinflammatory factors

and anti-inflammatory factors. In IBD, immune response activation

causes the release of inflammatory cytokines (eg, TNF-α) and

growth factors (eg, vascular endothelial growth factor [VEGF]) into

8

median levels of ICAM-1 were significantly higher in patients

with active CD vs controls (273 vs 168 ng/mL; P < .003). Median

circulating VCAM-1 concentrations were significantly higher in

active vs inactive UC patients (165 vs 117 U/mL; P < 005) and

were also significantly elevated compared with VCAM-1

concentrations in patients with active CD (124 U/mL; P < .02)

and in healthy controls (50 U/mL; P < .0001).77 It should be noted

that increased levels of ICAM and VCAM are not specific for

IBD and can be found in other inflammatory, infectious, and

neoplastic diseases.84

Vascular Endothelial Growth Factor

Angiogenesis, the growth of new blood vessels from preexisting

vasculature, is important in embryogenesis, tissue growth, wound

healing, and ovulation. Angiogenesis plays a key role in pathologic

processes as well, including cancer, ischemic cardiovascular

disease, diabetic retinopathy, and in chronic inflammatory diseases

such as IBD.85-87

VEGFs represent a family of factors that mediate angiogenesis.

Stimulation of angiogenesis is known to occur as a result of hypoxia

or mechanical shear stress and stretch. Inflammation that underlies

the pathology of IBD may promote angiogenesis in a number of

ways. Many inflammatory cytokines upregulated in IBD are

proangiogenic including IL-17 and TNF-α.88,89 Inflamed tissue,

such as that seen in IBD, is often hypoxic,90 and this hypoxia can

induce angiogenesis through upregulation of factors such as VEGF,

basic fibroblast growth factor-1, TNF-α, and hypoxia-inducible

factor-1.91,92 Neovascularization may also be stimulated by

extravasated fibrinogen.93 Angiogenic factors produced by

macrophages, mast cells, lymphocytes, and fibroblasts can stimulate

growth of blood vessels.91 Additionally, shear stress on the

endothelium related to increased blood flow itself may stimulate

angiogenesis.94,95 Several studies have demonstrated that circulating

levels of VEGF are elevated in patients with IBD compared with

healthy or disease controls and that VEGF levels correlate with

disease activity in IBD.96-101

C-Reactive Protein

CRP, predominantly produced in liver cells, is one of at least

40 proteins that participate acutely in the inflammatory response.

Normally, the amount of CRP produced by hepatocytes is low

(<1 mg/L), but following an acute stimulus such as inflammation, the

production of CRP is rapidly increased under the influence of IL-6,

TNF-α, and IL-1β.102 While the function of CRP in vivo is not

completely understood, CRP is a useful marker for detecting infections

and inflammation due to its rapid rise and short half-life (~19 h)

and for assessing the effect of therapy on the underlying disease,

as resolution of the stimulus triggering its production normalizes

CRP levels.102,103

Shine and colleagues were the first to show that a CRP

increase can be used to differentiate IBD from functional bowel

disorders. In 82 patients with chronic abdominal symptoms, 19

were diagnosed with CD, 22 with UC, and 41 with a functional

bowel disorder. All of the 19 patients with CD and 59% of the 22

patients with UC showed increases in CRP compared with none

of the 41 patients with functional symptoms.104 Schoepfer and

colleagues found that CRP had a 64% sensitivity and a 92%

specificity in discriminating IBD (n=64) from IBS (n=30).105 A much

larger study performed by Henriksen and colleagues in Norway

provided CRP data on 454 patients with UC and 200 patients

with CD, which was measured at diagnosis, with follow-up at 1

and 5 years.103 Patients with CD were noted to have a stronger mean

CRP response when compared with those with UC at diagnosis

(CD 51 mg/L vs UC 18 mg/L; P< .001), at 1 year (CD 51 mg/L vs

UC 18 mg/L; P< .001) and at 5 years (CD 13 mg/L vs UC 6 mg/L;

P< .001) after diagnosis. Additionally, these investigators found

that CRP levels at the time of diagnosis were related to the extent

of disease in patients with UC, but no such association was found

in CD.103

Serum Amyloid A

Serum amyloid A (SAA) is a family of four proteins, including

the acute phase proteins SAA1 and SAA2, whose primarily hepatic

expression is markedly increased in response to a variety of inflammatory

stimuli,106,107 much like CRP. As SAA has a physiologic level that is

10 times higher than that for CRP, it may allow for easier detection

of slight elevations.108 Its longer half-life (24 hours) compared with

that of CRP (19 hours) may make SAA more sensitive as an acute

phase reactant. In contrast to differential CRP levels that have been

seen in CD versus UC (see above), Niederau and colleagues found

that SAA levels were similarly elevated in patients with both forms

of IBD.109 Jijon and colleagues have demonstrated that SAA activates

the NF-κB signaling pathway, highlighting the proinflammatory

capacity of SAA.110 NF-κB is known to be one of the major regulatory

components involved in the dysregulation of cytokine production

and signaling mechanisms by intestinal epithelial cells, lymphocytes,

and macrophages that has been implicated in the pathogenesis

of IBD.111

A summary of the new serologic, genetic, and inflammatory

biomarkers that have been included in the new PROMETHEUS IBD sgi

Diagnostic test can be found in Table 1.

9

Table 1. New Serologic, Genetic, and Inflammation Biomarkers Included in PROMETHEUS IBD sgi Diagnostic Test

Serologic Markers

Marker General Prevalence Role/Function Effect

Anti-Fla-X and ● 57% of CD patients44

anti-A4-Fla2 (anti-Fla-X)

● 59% of CD patients44

(anti-A4-Fla2)

Genetic Markers

Marker General Prevalence Role/Function Effect

ATG16L1 ● OR 1.4550

associated

with CD

ECM1 ● OR 1.3–1.457

associated

with UC

NKX2.3 ● OR 1.2–1.657

associated with

CD and UC

STAT3 ● OR 1.1846

associated with

CD and UC

Inflammation Markers

Marker General Prevalence Role/Function Effect

VEGF ● Elevated in in some

patients with CD

and UC and in distal

colon tissue75

ICAM and ● Expressed in high

VCAM levels in IBD patients75

CRP ● Elevated in the context

of inflammation

● CRP response may be higher

in CD vs UC patients103

SAA ● More sensitive but less

specific than CRP in

acute-phase monitoring

of IBD113

● Fla-X38 and A4-Fla242 are flagellins

● Flagellin is the primary structural

component of bacterial flagella,

used for motility

● Serve as immunodominant antigens in

the microbiota38

● Valuable for the diagnosis of IBD,

especially for the more precise

discrimination of CD from UC

● Elongates membranes that form

autophagosomes in autophagy50

● Interacts with basal membrane, inhibits

MMP9 proteins, and activates signaling of

proinflammatory cytokine pathway NF-κB46

● Encodes transcription factor that

functions in cell specification and

maintenance of tissue differentiation46

● Plays a role in the Th17-dependent

autoimmune process

● May decrease ability to remove

intestinal microbes51

● Increases production of inflammatory

cytokines from macrophages51

● Exhibits defective barrier function46

● Causes thickening and scarring of

mucus membranes68

● Associated with defects in gut

development, abnormal tissue

architecture, abnormalities in migration

and segregation of B and T cells57

● Expansion and overactivity of Th17

helper cell cytokines can lead to

intestinal inflammation112

● Proangiogenic factor that causes

vasodilation through nitric oxide release

and increased permeability, endothelial

cell proliferation, and directed migration

and differentiation75

● Promotes recruitment of inflammatory cells76

● Involved in slow rolling of lymphocytes

and monocytes during inflammatory cell

recruitment75

● Acute phase reactant that increases

markedly when stimulated by

inflammation102

● Promotes leukocyte chemotaxis,

cellular adhesion

● Increases cytokine production and

metalloproteinase secretion in various

cell types110

● Extensive transmural injury including

edema, loss of goblet cells, decreased

mucus production, crypt cell hyperplasia,

and erosions and ulcerations75

● Initiation of leukocyte migration and

local tissue inflammation77

● Circulating levels correlate well with

disease activity77

● Can be used to differentiate IBD

from functional bowel disorders104

● Levels correlate with extent of

disease in UC103

● May directly influence the

development of inflammation in

the intestine110

10

Figure 5. The corresponding positive and negative predictive values

(PPV and NPV, respectively) are listed in Table 3. When IBD is highly

suspected, the PPV with the PROMETHEUS IBD sgi Diagnostic test

is 98%. When IBD suspicion is low, the NPV with the PROMETHEUS

IBD sgi Diagnostic test is 95%.

Figure 4. Receiver Operator Characteristics (All Individual Markers)

1-Specificity (False-Positive Rate)

IBD vs non-IBD: All Continuous Markers

Sens

itiv

ity

(Tru

e-Po

sitive

Rat

e)

1

0.9

0.8

0.7

0.6

0.5

0.4

0.3

0.2

0.1

00 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 1

AUCTest0.690

0.606

0.653

0.642

0.640

0.624

0.638

0.610

0.653

0.581

0.563

0.651

0.500

ASCA-IgA

ASCA-IgG

ANCA

OmpC

CBir1

A4-Fla2

Fla-X

CRP

SAA

ICAM

VCAM

VEGF

No Discrimination

Table 2. Patient Allocation for the Training and Validation Phases

of Development of the PROMETHEUS IBD sgi Diagnostic Test

Disease Training Cohort Validation Cohort

Diagnosis n (%) n (%)

Crohn’s Disease 419 (39) 153 (35)

Ulcerative Colitis 227 (21) 101 (23)

GI Controls 314 (29) 123 (28)

Healthy Controls 123 (11) 60 (14)

Total 1083 (100) 437 (100)

Development and Validation of the Prometheus IBD sgi Diagnostic Test

The PROMETHEUS IBD sgi Diagnostic Algorithm

The IBD sgi Diagnostic test algorithm was developed using a

computational method called “Random Forest.”

The algorithm was developed using a total of 1,520 well-

characterized samples from IBD and non-IBD patients from 50 centers

in North America. A total of 1,083 samples were used to train the

algorithm, and an independent cohort of 437 samples was used to

validate and determine the performance of the test.

The algorithm uses measurements of 17 biologic markers (blood

proteins and genes) to first predict whether a patient has IBD. Then, if

the patient is predicted to have IBD, the algorithm examines the

marker measurements to differentiate CD from UC.

The training and validation cohorts used to develop the

PROMETHEUS IBD sgi Diagnostic test are detailed below in Table 2.

“GI controls” were subjects who had non-IBD gastrointestinal disease.

Receiver Operator Characteristics (ROC) Analysis

The accuracy of any diagnostic test is dependent on how well the

test separates patients with a specified condition versus those without

the condition. In Figure 4, the ROC curves corresponding to each

individual marker of the new PROMETHEUS IBD sgi Diagnostic test

are presented. In this figure, AUC represents the performance of the

test. If an AUC value of 1 is obtained, this is representative of a

“perfect” test to discriminate between those with and those without

disease. An AUC of 0.5 represents a discriminatory performance that

is equal to that achieved with random chance.

When the patterns of all biomarkers are combined and

examined by the PROMETHEUS IBD sgi Diagnostic test algorithm, the

performance of the test improved significantly compared with the

performances of the individual markers alone, as illustrated in

Figure 5. Receiver Operator Characteristics: PROMETHEUS IBD sgi

Diagnostic Test

1-Specificity (False-Positive Rate)

Sens

itiv

ity

(Tru

e-Po

sitive

Rat

e)

1

0.9

0.8

0.7

0.6

0.5

0.4

0.3

0.2

0.1

00 1

AUCTest0.871sgi

AUCTest

0.690

0.606

0.653

0.642

0.640

0.624

0.638

0.610

0.653

0.581

0.563

0.651

0.500

ASCA-IgA

ASCA-IgG

ANCA

OmpC

CBir1

A4-Fla2

Fla-X

CRP

SAA

ICAM

VCAM

VEGF

No Discrimination

IBD vs non-IBD: All Continuous Markers

0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 1

11

Table 3. Positive and Negative Predictive Values for the

PROMETHEUS IBD sgi Diagnostic Test

Prevalence, % IBD, %

Positive 15 56

predictive value (PPV)

50 88

85 98

Negative 15 95

predictive value (NPV)

50 77

85 37

Conclusion

The diagnosis of IBD and the differentiation between CD and UC

can pose a number of challenges in the clinical setting. Invasive

radiographic and endoscopic diagnostic investigations are not

without complications, and there is a subset of patients for whom

these tests do not result in a definitive diagnosis. Diagnostic delays

may adversely affect the management of IBD and illustrate the need for

diagnostic accuracy. Ongoing research into the underlying

pathophysiology of IBD has helped to identify more markers,

including serologic, genetic, and inflammatory markers, that may

prove to be a useful, noninvasive adjunctive diagnostic tool when IBD

is suspected. Helping to provide actionable diagnostic information

has the potential to allow for personalized treatment planning and

better patient outcomes.

12

13

18. Russell RK, Satsangi J. IBD: a family affair. Best Pract Res Clin

Gastroenterol. 2004;18(3):525-539.

19. Human genome project information: SNP fact sheet. U.S.

Department of Energy Genome Program. Available at: http://

www.ornl.gov/sci/techresources/Human_Genome/faq/snps.

shtml. Accessed: September 27, 2011.

20. Seidman EG. Emerging prognostic markers in IBD. Gastroenterol

Hepatol. 2007;3(12 Suppl 36):5-6.

21. Sendid B, Colombel JF, Jacquinot PM, et al. Specific antibody

response to oligomannosidic epitopes in Crohn’s disease. Clin

Diagn Lab Immunol. 1996;3(2):219-226.

22. Vasiliauskas EA, Plevy SE, Landers CJ, et al. Perinuclear

antineutrophil cytoplasmic antibodies in patients with Crohn’s

disease define a clinical subgroup. Gastroenterology. 1996;110(6):

1810-1819.

23. Reese GE, Constantinides VA, Simillis C, et al. Diagnostic precision

of anti-Saccharomyces cerevisiae antibodies and perinuclear

antineutrophil cytoplasmic antibodies in inflammatory bowel

disease. Am J Gastroenterol. 2006;101(10):2410-2422.

24. Joossens S, Reinisch W, Vermeire S, et al. The value of serologic

markers in indeterminate colitis: a prospective follow-up study.

Gastroenterology. 2002;122(5):1242-1247.

25. Dubinsky M. What is the role of serological markers in IBD?

Pediatric and adult data. Dig Dis. 2009;27(3):259-268.

26. Dubinsky MC, Hanauer SB, [section editor]. Emerging strategies in

the use of IBD-related serologic markers (in Advances in IBD [Q&A

section]). Gastroenterology & Hepatology. 2008;4(8):1-3.

27. Standaert-Vitse A, Jouault T, Vandewalle P, et al. Candida albicans

is an immunogen for anti-Saccharomyces cerevisiae antibody

markers of Crohn’s disease. Gastroenterology. 2006;130(6):

1764-1775.

28. Standaert-Vitse A, Sendid B, Joossens M, et al. Candida albicans

colonization and ASCA in familial Crohn’s disease. Am J

Gastroenterol. 2009;104(7):1745-1753.

29. Sandborn WJ. Serologic markers in inflammatory bowel disease:

state of the art. Rev Gastroenterol Disord. 2004;4(4):167-174.

30. Peyrin-Biroulet L, Standaert-Vitse A, Branche J, Chamaillard M.

IBD serological panels: facts and perspectives. Inflamm Bowel Dis.

2007;13(12):1561-1566.

31. Cohavy O, Bruckner D, Gordon LK, et al. Colonic bacteria express

an ulcerative colitis pANCA-related protein epitope. Infect Immun.

2000;68(3):1542-1548.

32. Arnott ID, Landers CJ, Nimmo EJ, et al. Sero-reactivity to microbial

components in Crohn’s disease is associated with disease severity

and progression, but not NOD2/CARD15 genotype. Am J

Gastroenterol. 2004;99(12):2376-2384.

33. Landers CJ, Cohavy O, Misra R, et al. Selected loss of tolerance

evidenced by Crohn’s disease-associated immune responses to auto-

and microbial antigens. Gastroenterology. 2002;123(3):689-699.

34. Mow WS, Vasiliauskas EA, Lin YC, et al. Association of antibody

responses to microbial antigens and complications of small bowel

Crohn’s disease. Gastroenterology. 2004;126(2):414-424.

References

01. Matricon J, Barnich N, Ardid D. Immunopathogenesis of

inflammatory bowel disease. Self Nonself. 2010;1(4):299-309.

02. Podolsky DK. Inflammatory bowel disease. N Engl J Med. 2002;

347(6):417-429.

03. Xavier RJ, Podolsky DK. Unravelling the pathogenesis of

inflammatory bowel disease. Nature. 2007;448(7152):427-434.

04. Sands BE. From symptom to diagnosis: clinical distinctions among

various forms of intestinal inflammation. Gastroenterology. 2004;

126(6):1518-1532.

05. Ruemmele FM, Targan SR, Levy G, et al. Diagnostic accuracy of

serological assays in pediatric inflammatory bowel disease.

Gastroenterology. 1998;115(4):822-829.

06. Seidman EG. Recent advances in the diagnosis and treatment of

pediatric inflammatory bowel disease. Curr Gastroenterol Rep.

2000;2(3):248-252.

07. Lichtenstein GR, Hanauer SB, Sandborn WJ, and the Practice

Parameters Committee of the American College of Gastroenterology.

Management of Crohn’s disease in adults. Am J Gastroenterol.

2009;104(2):465-483.

08. Nikolaus S, Schreiber S. Diagnostics of inflammatory bowel

disease. Gastroenterology. 2007;133(5):1670-1689.

09. Fletcher JG, Fidler JL, Bruining DH, Huprich JE. New concepts in

intestinal imaging for inflammatory bowel diseases. Gastroenterology.

2011;140(6):1795-1806.

10. Cheifetz AS, Kornbluth AA, Legnani P, et al. The risk of retention of

the capsule endoscope in patients with known or suspected Crohn’s

disease. Am J Gastroenterol. 2006;101(10):2218-2222.

11. Lohsiriwat V. Colonoscopic perforation: incidence, risk factors,

management and outcome. World J Gastroenterol. 2010;16(4):

425-430.

12. Vavricka SR, Spigaglia SM, Rogler G, et al. Systematic evaluation of

risk factors for diagnostic delay in inflammatory bowel disease.

Inflamm Bowel Dis. 2011; epub ahead of print.

13. Pezim ME, Pemberton JH, Beart RW, Jr., et al. Outcome of

"indeterminant" colitis following ileal pouch-anal anastomosis. Dis

Colon Rectum. 1989;32(8):653-658.

14. Wolfson DM, Sachar DB, Cohen A, et al. Granulomas do not affect

postoperative recurrence rates in Crohn’s disease. Gastroenterology.

1982;83(2):405-409.

15. Papadakis KA, Tabibzadeh S. Diagnosis and misdiagnosis of

inflammatory bowel disease. Gastrointest Endosc Clin North Am.

2002;12(3):433-449.

16. Orholm M, Binder V, Sorensen TI, et al. Concordance of inflammatory

bowel disease among Danish twins. Results of a nationwide study.

Scand J Gastroenterol. 2000;35(10):1075-1081.

17. Tysk C, Lindberg E, Jarnerot G, Floderus-Myrhed B. Ulcerative

colitis and Crohn’s disease in an unselected population of

monozygotic and dizygotic twins. A study of heritability and the

influence of smoking. Gut. 1988;29(7):990-996.

14

35. Markowitz J, Kugathasan S, Dubinsky M, et al. Age of diagnosis

influences serologic responses in children with Crohn’s disease: a

possible clue to etiology? Inflamm Bowel Dis. 2009;15(5):714-719.

36. Zholudev A, Zurakowski D, Young W, et al. Serologic testing with

ANCA, ASCA, and anti-OmpC in children and young adults with

Crohn’s disease and ulcerative colitis: diagnostic value and

correlation with disease phenotype. Am J Gastroenterol. 2004;

99(11):2235-2241.

37. Zeng H, Carlson AQ, Guo Y, et al. Flagellin is the major

proinflammatory determinant of enteropathogenic Salmonella.

J Immunol. 2003;171(7):3668-3674.

38. Lodes MJ, Cong Y, Elson CO, et al. Bacterial flagellin is a dominant

antigen in Crohn disease. J Clin Invest. 2004;113(9):1296-1306.

39. Sitaraman SV, Klapproth JM, Moore DA, III, et al. Elevated flagellin-

specific immunoglobulins in Crohn’s disease. Am J Physiol

Gastrointest Liver Physiol. 2005;288(2):G403-G406.

40. Targan SR, Landers CJ, Yang H, et al. Antibodies to CBir1 flagellin

define a unique response that is associated independently with

complicated Crohn’s disease. Gastroenterology. 2005;128(7):

2020-2028.

41. Collins MD, Lawson PA, Willems A, et al. The phylogeny of the

genus Clostridium: proposal of five new genera and eleven new

species combinations. Int J Syst Bacteriol. 1994;44(4):812-826.

42. Duck LW, Walter MR, Novak J, et al. Isolation of flagellated bacteria

implicated in Crohn’s disease. Inflamm Bowel Dis. 2007;13(10):

1191-1201.

43. Schoepfer AM, Schaffer T, Seibold-Schmid B, et al. Antibodies to

flagellin indicate reactivity to bacterial antigens in IBS patients.

Neurogastroenterol Motil. 2008;20:1110-1118.

44. Schoepfer AM, Schaffer T, Mueller S, et al. Phenotypic associations

of Crohn’s disease with antibodies to flagellins A4-Fla2 and Fla-

X, ASCA, p-ANCA, PAB, and NOD2 mutations in a Swiss Cohort.

Inflamm Bowel Dis. 2009;15(9):1358-1367.

45. Vidrich A, Lee J, James E, et al. Segregation of pANCA antigenic

recognition by DNase treatment of neutrophils: ulcerative colitis,

type 1 autoimmune hepatitis, and primary sclerosing cholangitis.

J Clin Immunol. 1995;15(6):293-299.

46. Lees CW, Satsangi J. Genetics of inflammatory bowel disease:

implications for disease pathogenesis and natural history. Expert

Rev Gastroenterol Hepatol. 2009;3(5):513-534.

47. Deretic V. Links between autophagy, innate immunity, inflammation

and Crohn’s disease. Dig Dis. 2009;27(3):246-251.

48. Hampe J, Franke A, Rosenstiel P, et al. A genome-wide association

scan of nonsynonymous SNPs identifies a susceptibility variant for

Crohn disease in ATG16L1. Nat Genet. 2007;39(2):207-211.

49. SNP class definition. National Center for Biotechnology

Information; U.S. National Library of Medicine. Available at:

http://www.ncbi.nlm.nih.gov/books/NBK44488/. Accessed:

September 27, 2011.

50. Cummings JR, Cooney R, Pathan S, et al. Confirmation of the role

of ATG16L1 as a Crohn’s disease susceptibility gene. Inflamm

Bowel Dis. 2007;13(8):941-946.

51. Cheng JF, Ning YJ, Zhang W, et al. T300A polymorphism of

ATG16L1 and susceptibility to inflammatory bowel diseases: a

meta-analysis. World J Gastroenterol. 2010;16(10):1258-1266.

52. Palomino-Morales RJ, Oliver J, Gomez-Garcia M, et al. Association

of ATG16L1 and IRGM genes polymorphisms with inflammatory

bowel disease: a meta-analysis approach. Genes Immun.

2009;10(4):356-364.

53. Wellcome Trust Case Control Consortium. Genome-wide association

study of 14,000 cases of seven common diseases and 3,000 shared

controls. Nature. 2007;447(7145):661-678.

54. Yamazaki K, Takahashi A, Takazoe M, et al. Positive association of

genetic variants in the upstream region of NKX2-3 with Crohn’s

disease in Japanese patients. Gut. 2009;58(2):228-232.

55. Weersma RK, Stokkers PC, Cleynen I, et al. Confirmation of multiple

Crohn’s disease susceptibility loci in a large Dutch-Belgian cohort.

Am J Gastroenterol. 2009;104(3):630-638.

56. Fisher SA, Tremelling M, Anderson CA, et al. Genetic determinants

of ulcerative colitis include the ECM1 locus and five loci implicated

in Crohn’s disease. Nat Genet. 2008;40(6):710-712.

57. Meggyesi N, Kiss LS, Koszarska M, et al. NKX2-3 and IRGM

variants are associated with disease susceptibility to IBD in Eastern

European patients. World J Gastroenterol. 2010;16(41):5233-5240.

58. Wegenka UM, Buschmann J, Lutticken C, et al. Acute-phase

response factor, a nuclear factor binding to acute-phase response

elements, is rapidly activated by interleukin-6 at the posttranslational

level. Mol Cell Biol. 1993;13(1):276-288.

59. Rawlings JS, Rosler KM, Harrison DA. The JAK/STAT signaling

pathway. J Cell Sci. 2004;117(Pt 8):1281-1283.

60. Takeda K, Clausen BE, Kaisho T, et al. Enhanced Th1 activity and

development of chronic enterocolitis in mice devoid of Stat3 in

macrophages and neutrophils. Immunity. 1999;10(1):39-49.

61. Wolk K, Kunz S, Witte E, et al. IL-22 increases the innate immunity

of tissues. Immunity. 2004;21(2):241-254.

62. Yasukawa H, Sasaki A, Yoshimura A. Negative regulation of cytokine

signaling pathways. Annu Rev Immunol. 2000;18:143-164.

63. Adamson AS, Collins K, Laurence A, O’Shea JJ. The Current STATus

of lymphocyte signaling: new roles for old players. Curr Opin

Immunol. 2009;21(2):161-166.

64. Chen Z, Laurence A, O’Shea JJ. Signal transduction pathways and

transcriptional regulation in the control of Th17 differentiation.

Semin Immunol. 2007;19(6):400-408.

65. Sugimoto K. Role of STAT3 in inflammatory bowel disease. World J

Gastroenterol. 2008;14(33):5110-5114.

66. Barrett JC, Hansoul S, Nicolae DL, et al. Genome-wide association

defines more than 30 distinct susceptibility loci for Crohn’s disease.

Nat Genet. 2008;40(8):955-962.

67. Cenit MC, Alcina A, Marquez A, et al. STAT3 locus in inflammatory

bowel disease and multiple sclerosis susceptibility. Genes Immun.

2010;11(3):264-268.

68. Thompson AI, Lees CW. Genetics of ulcerative colitis. Inflamm

Bowel Dis. 2011;17(3):831-848.

15

086. Carmeliet P. Angiogenesis in health and disease. Nat Med. 2003;

9(6):653-660.

087. Carmeliet P. Angiogenesis in life, disease and medicine. Nature.

2005;438(7070):932-936.

088. Polzer K, Baeten D, Soleiman A, et al. Tumour necrosis factor

blockade increases lymphangiogenesis in murine and human

arthritic joints. Ann Rheum Dis. 2008;67(11):1610-1616.

089. Heidenreich R, Rocken M, Ghoreschi K. Angiogenesis drives

psoriasis pathogenesis. Int J Exp Pathol. 2009;90(3):232-248.

090. Eltzschig HK, Carmeliet P. Hypoxia and inflammation. N Engl J

Med. 2011;364(7):656-665.

091. Qu Z, Liebler JM, Powers MR, et al. Mast cells are a major source

of basic fibroblast growth factor in chronic inflammation and

cutaneous hemangioma. Am J Pathol. 1995;147(3):564-573.

092. Taylor PC, Sivakumar B. Hypoxia and angiogenesis in rheumatoid

arthritis. Curr Opin Rheumatol. 2005;17(3):293-298.

093. Hatton MW, Southward SM, Legault KJ, et al. Fibrinogen catabolism

within the procoagulant VX-2 tumor of rabbit lung in vivo:

Effluxing fibrin(ogen) fragments contain antiangiogenic activity.

J Lab Clin Med. 2004;143(4):241-254.

094. Cullen JP, Sayeed S, Sawai RS, et al. Pulsatile flow-induced

angiogenesis: role of G(i) subunits. Arterioscler Thromb Vasc Biol.

2002;22(10):1610-1616.

095. Schaper W, Scholz D. Factors regulating arteriogenesis. Arterioscler

Thromb Vasc Biol. 2003;23(7):1143-1151.

096. Bousvaros A, Leichtner A, Zurakowski D, et al. Elevated serum

vascular endothelial growth factor in children and young adults

with Crohn’s disease. Dig Dis Sci. 1999;44(2):424-430.

097. Di Sabatino A, Ciccocioppo R, Armellini E, et al. Serum bFGF and

VEGF correlate respectively with bowel wall thickness and

intramural blood flow in Crohn’s disease. Inflamm Bowel Dis.

2004;10(5):573-577.

098. Duenas Pousa I, Mate Jimenez J, Salcedo Mora X, et al. Analysis

of soluble angiogenic factors in Crohn’s disease: a preliminary

study. Gastroenterol Hepatol. 2007;30(9):518-524.

099. Griga T, Tromm A, Spranger J, May B. Increased serum levels of

vascular endothelial growth factor in patients with inflammatory

bowel disease. Scand J Gastroenterol. 1998;33(5):504-508.

100. Kanazawa S, Tsunoda T, Onuma E, et al. VEGF, basic-FGF, and

TGF-beta in Crohn’s disease and ulcerative colitis: a novel

mechanism of chronic intestinal inflammation. Am J Gastroenterol.

2001;96(3):822-828.

101. Pousa ID, Mate J, Salcedo-Mora X, et al. Role of vascular

endothelial growth factor and angiopoietin systems in serum of

Crohn’s disease patients. Inflamm Bowel Dis. 2008;14(1):61-67.

102. Vermeire S, Van Assche G, Rutgeerts P. Laboratory markers in IBD:

useful, magic, or unnecessary toys? Gut. 2006;55(3):426-431.

103. Henriksen M, Jahnsen J, Lygren I, et al. C-reactive protein: a

predictive factor and marker of inflammation in inflammatory

bowel disease. Results from a prospective population-based study.

Gut. 2008;57(11):1518-1523.

69. Arnott ID, Kingstone K, Ghosh S. Abnormal intestinal permeability

predicts relapse in inactive Crohn disease. Scand J Gastroenterol.

2000;35(11):1163-1169.

70. Wyatt J, Vogelsang H, Hubl W, et al. Intestinal permeability and the

prediction of relapse in Crohn’s disease. Lancet. 1993;341(8858):

1437-1439.

71. Matsuda A, Suzuki Y, Honda G, et al. Large-scale identification and

characterization of human genes that activate NF-κB and MAPK

signaling pathways. Oncogene. 2003;22(21):3307-3318.

72. Wang L, Yu J, Ni J, et al. Extracellular matrix protein 1 (ECM1) is

over-expressed in malignant epithelial tumors. Cancer Lett.

2003;200(1):57-67.

73. Anderson CA, Massey DC, Barrett JC, et al. Investigation of Crohn’s

disease risk loci in ulcerative colitis further defines their molecular

relationship. Gastroenterology. 2009;136(2):523-529.

74. Sands BE, Kaplan GG. The role of TNF-α in ulcerative colitis. J

Clin Pharmacol. 2007;47(8):930-941.

75. Chidlow JH, Jr., Shukla D, Grisham MB, Kevil CG. Pathogenic

angiogenesis in IBD and experimental colitis: new ideas and

therapeutic avenues. Am J Physiol Gastrointest Liver Physiol.

2007;293(1):G5-G18.

76. Rijcken E, Krieglstein CF, Anthoni C, et al. ICAM-1 and VCAM-1

antisense oligonucleotides attenuate in vivo leucocyte adherence

and inflammation in rat inflammatory bowel disease. Gut.

2002;51(4):529-535.

77. Jones SC, Banks RE, Haidar A, et al. Adhesion molecules in

inflammatory bowel disease. Gut. 1995;36(5):724-730.

78. Simmons D, Makgoba MW, Seed B. ICAM, an adhesion ligand of

LFA-1, is homologous to the neural cell adhesion molecule NCAM.

Nature. 1988;331(6157):624-627.

79. Myers CL, Wertheimer SJ, Schembri-King J, et al. Induction of

ICAM-1 by TNF-α, IL-1β, and LPS in human endothelial cells after

downregulation of PKC. Am J Physiol. 1992;263(4 Pt 1):

C767-C772.

80. Haraldsen G, Kvale D, Lien B, et al. Cytokine-regulated expression

of E-selectin, intercellular adhesion molecule-1 (ICAM-1), and

vascular cell adhesion molecule-1 (VCAM-1) in human microvascular

endothelial cells. J Immunol. 1996;156(7):2558-2565.

81. Perry MA, Phillipson M, Holm L. Transmural gradient of leukocyte-

endothelial interaction in the rat gastrointestinal tract. Am J Physiol

Gastrointest Liver Physiol. 2005;289(5):G852-G859.

82. Shimizu Y, Newman W, Tanaka Y, Shaw S. Lymphocyte

interactions with endothelial cells. Immunol Today. 1992;13(3):

106-112.

83. Panes J, Granger DN. Leukocyte-endothelial cell interactions:

molecular mechanisms and implications in gastrointestinal disease.

Gastroenterology. 1998;114(5):1066-1090.

84. Mustjoki S, Alitalo R, Elonen E, et al. Intercellular adhesion

molecule-1 in extravasation of normal mononuclear and leukaemia

cells. Br J Haematol. 2001;113(4):989-1000.

85. Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other

disease. Nat Med. 1995;1(1):27-31.

16

104. Shine B, Berghouse L, Jones JE, Landon J. C-reactive protein as

an aid in the differentiation of functional and inflammatory bowel

disorders. Clin Chim Acta. 1985;148(2):105-109.

105. Schoepfer AM, Trummler M, Seeholzer P, et al. Discriminating IBD

from IBS: comparison of the test performance of fecal markers,

blood leukocytes, CRP, and IBD antibodies. Inflamm Bowel Dis.

2008;14(1):32-39.

106. Malle E, de Beer FC. Human serum amyloid A (SAA) protein: a

prominent acute-phase reactant for clinical practice. Eur J Clin

Invest. 1996;26(6):427-435.

107. Uhlar CM, Whitehead AS. Serum amyloid A, the major vertebrate

acute-phase reactant. Eur J Biochem. 1999;265(2):501-523.

108. Yamada T. Serum amyloid A (SAA): a concise review of biology,

assay methods and clinical usefulness. Clin Chem Lab Med. 1999;

37(4):381-388.

109. Niederau C, Backmerhoff F, Schumacher B, Niederau C.

Inflammatory mediators and acute phase proteins in patients with

Crohn’s disease and ulcerative colitis. Hepatogastroenterology.

1997;44(13):90-107.

110. Jijon HB, Madsen KL, Walker JW, et al. Serum amyloid A activates

NF-κB and proinflammatory gene expression in human and murine

intestinal epithelial cells. Eur J Immunol. 2005;35(3):718-726.

111. Atreya I, Atreya R, Neurath MF. NF-∫B in inflammatory bowel

disease. J Intern Med. 2008;263(6):591-596.

112. Abraham C, Cho JH. Inflammatory bowel disease. N Engl J Med.

2009;361(21):2066-2078.

113. Chambers RE, Stross P, Barry RE, Whicher JT. Serum amyloid

A protein compared with C-reactive protein, alpha 1-antichymotrypsin

and alpha 1-acid glycoprotein as a monitor of inflammatory bowel

disease. Eur J Clin Invest. 1987;17(5):460-467.

INSIDE BACK COVER(BLANK)

9410 Carroll Park Drive

San Diego, CA 92121

888-423-5227

858-824-0896 fax

www.prometheuslabs.com

Prometheus diagnostic services provide important information to aid in the diagnosis

and management of certain diseases and conditions.

How this information is used to guide patient care is the responsibility of the physician.

PROMETHEUS and the Link Design are trademarks or registered trademarks of Prometheus Laboratories Inc.

© 2011 Prometheus Laboratories Inc. All rights reserved. CD10037 09/11

Prometheus products, services, and technology are covered by one or more

US patents and patents pending. For more information, see www.prometheuslabs.com.