You are on page 1of 96

Consensus statements from Saudi Ministry of Health regarding the diagnosis and

treatment of inflammatory bowel disease (IBD)

ABSTRACT:

Background: The term inflammatory bowel disease (IBD) encompasses two clinical entities:

ulcerative colitis (UC) and Crohn’s disease (CD), which reflectreflects the characteristic of

chronic inflammation of the gastrointestinal tract, and display heterogennicheterogenic

clinical manifestations between patients, and within individuals over time. Optimal

management relies on understanding and tailoring evidence-based interventions by clinicians

in partnership with patients.

Aim and Sscope: We aimed to provide a condensed guideline of IBD management for

different categories of patients, which could be used as a tool by clinicians in order to

develop an individualized patient’s management plan. The current recommendations are

based on the most up-to-date information at the time of writing, and will be updated on a

regular basis.

Furthermore, the current recommendations are not intended to be used as rigid therapeutical

guidelines. They are also not meant to take the place of practicing physician’s clinical

judgement; instead, they are meant to assist and advise health care practitioners who are

managing IBD patients.

Materials and Mmethods: Theseis guidelines for the management of inflammatory bowel

diseaseIBD in adults and children was developed by the Saudi Ministry of Health in

collaboration with the Saudi gastroenterology association (SGA) and Saudi society of clinical

pharmacology (SCCP).national specialists.

1
A literature review of the current publications, and international guidelines regarding the

management of IBD was performed. A total of 145 evidence and expert opinion-based

recommendations for diagnosis and treatment of UC and CD were proposed, and further

refined by a voting process. Participants suggested revisions and commented on the

statements, after which, the specific statements were revised.

The quality of evidence was considered:

– High, if further research very unlikely to change confidence in the estimate of effect;

– Moderate, if further research likely to have an important impact on confidence in the

estimate of effect, and may change the estimate;

– Low, if further research very likely to have an important impact on confidence in the

estimate of effect, and is likely to change the estimate;

A summary of the current IBD treatment goals and algorithms can be consulted as a

supplementary material (Supplement 1).

Results: The consensus includes 145 statements focused on diagnosis and medical treatment

options of IBD.

The group supported the use of serological biochemical markers (i.e., fecal calprotectin), ileo-

colonoscopy with multiple biopsies, and cross-sectional imaging as important tools for a

definitive diagnosis. Moreover, several international scoring systems were proposed in order

to classify different phenotypes of IBD among various age groups.

The group reached agreement upon the positioning of corticosteroids therapies, exclusive

enteral nutrition, biologic agents, and 5-aminosalicylates (5-ASA), immunosuppressants and

biologic agents, as important treatment options of for IBD in (both UC and CD). Based on

consensus, the treat-to-target (TTT) strategy was chosen as the core treatment strategy for

IBD. The majority of the statements were backed up by high-quality evidence.

2
Conclusions:

A definitive diagnosis of UC or CD requires a multidisciplinary approach based on clinical

manifestations, serologic, endoscopic, imagingstic, and histologic factorsfeatures. The main

differential diagnoses should be made with enteric infections, intestinal TB, or primary

immune deficiency.

Evidence-based medical treatment of IBD in all patient categories is recommended, with

thorough ongoing assessments to define treatment success.

Table of Contents
Chapter 1. Diagnosis of Crohn’s disease (CD) and ulcerative colitis (UC).........................................................3
General considerations.....................................................................................................................................3
Ulcerative colitis Diagnosis...............................................................................................................................8
Crohn’s disease Diagnosis................................................................................................................................9
Classification of inflammatory bowel disease...............................................................................................12
Composite clinical and endoscopic disease activity in UC...........................................................................15
Endoscopic disease activity in UC..................................................................................................................16
The Pediatric Crohn’s Disease Activity Index (PDCAI).............................................................................18
Treatment Goals..............................................................................................................................................18
Treatment Algorithm......................................................................................................................................25
Crohn’s disease................................................................................................................................................26
Perianal Fistulizing Disease............................................................................................................................30
Ulcerative Colitis.............................................................................................................................................31
Biologics............................................................................................................................................................36
The use of biologics in moderate to severe UC.............................................................................................36
The use of biologics in moderate to severe CD.............................................................................................36
Sequencing of medications..............................................................................................................................40
Interventions based on therapeutic drug monitoring for patients with IBD and loss of response to anti-
TNF agents.......................................................................................................................................................41
Chapter 2. Special Patient population.................................................................................................................45
Using of biologics in pregnancy......................................................................................................................45
Diagnostic workup of children with suspected IBD.....................................................................................50

3
Medical Management of UC...........................................................................................................................55
ASA and Enemas.............................................................................................................................................55
Oral Corticosteroids........................................................................................................................................56
Immunosuppressants......................................................................................................................................57
Biologics............................................................................................................................................................58
General considerations for using Biologics...................................................................................................60
Medical management of Crohn’s disease in pediatrics (CD)......................................................................62
Aminosalicylates..............................................................................................................................................62
Corticosteroids.................................................................................................................................................63
Exclusive Enteral Nutrition............................................................................................................................64
Immunosuppressants......................................................................................................................................65
Anti–Tumor Necrosis Factor Biologic Therapy...........................................................................................66
References:.......................................................................................................................................................70

Chapter 1. Diagnosis of Crohn’s disease (CD) and ulcerative colitis (UC).


General considerations:

Statement 1: There are no precise diagnostic criteria for diagnosing Crohn’s disease (CD) or

ulcerative colitis (UC). A combination of clinical, biochemical, endoscopic, radiographic, and

histologic criteria is used to diagnose CD or UC.

Vote: 100% strongly agreed; high-quality evidence

Inflammatory bowel disease (IBD) includes a variety of intestinal disorders with an

unclear etiopathology, but common clinical manifestations. The most representative entities

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

Ulcerative colitisUC is a chronic inflammatory disease characterized by mucosal

inflammation starting distally in the rectum, with continuous extension proximally for a

variable distance, often with an abrupt demarcation between inflamed and non-inflamed

mucosa (1), while Crohn’s diseaseCD is a complex chronic inflammatory gastrointestinal

condition with variable age of onset, disease location, and behavior (2).

The diagnosis of IBD is not restricted to a certain criterion; however, the diagnosis is

based on a series of clinical, biochemical, endoscopic, radiographic, and histologic

4
parameters (3). Genetic and serological testing are not recommended as there is limited

evidence supporting their role in confirming IBD diagnosis (4).A

Although the diagnosis of IBD can be challenging, some of the presenting symptoms

may raise the suspicion of IBD. These may include hematochezia, diarrhea, tenesmus, and

abdominal cramping (5). Some enteric infections such as E. coli, Salmonella, Shigella,

Yersinia, and Campylobacter, and certain parasitic infections, such as amebiasis, may present

with similar symptoms, therefore it must be excluded before a final diagnosis of IBD is made

(6).

Statement 2: We recommend using the Red Flags Score (RFS) to help differentiate irritable

bowel syndrome (IBS) from inflammatory bowel disease (IBD).

Voting: 50% strongly agreed; high-quality evidence

The Red Flags Score (RFS) is a screening method developed to identify patients with

who are at a higher risk of having IBD rather than IBS (7). Danese et al., developed a 21-

items survey which was administered to healthy subjects, patients with irritable bowel

syndromeIBS (non-CD group) and patients with recently diagnosed (<18 months) CD (7).

The authors concluded that a minimum Red Flags indexRFS value of 8 was highly predictive

of CD diagnosis with sensitivity and specificity bootstrap estimates of 0.94 (95% confidence

interval 0.88–0.99) and 0.94 (0.90–0.97), respectively. Moreover, the association between

CD diagnosis and a Red Flags indexRFS value of ≥8 corresponded to an OR of 290 (p <

0.0001) in the above-mentioned study.

A more recent study, from Saudi Arabia, outlined the association between an elevated

RFS and the lack of specialized gastroenterological evaluation, thus appealing for early

specialized referrals (8).

5
Statement 3: Genetic and serological tests are not currently recommended for the

diagnosis of CD or UC. Complementary studies should focus on risk stratification and

disease activity evaluation at the time of diagnosis.

Vote: 100% strongly agreed; moderate-quality evidence

Genetic and serological testing are not recommended as there is limited evidence

supporting their role in confirming IBD diagnosis (4). Serological markers could be used in

order to support a diagnosis of IBD, though the accuracy of the best available tests is rather

limited and hence ineffective at differentiating colonic CD from UC (9).

Reese et al., conducted a meta-analysis to evaluate the diagnostic precision of anti-

Saccharomyces cerevisiae (ASCA) and perinuclear antineutrophil cytoplasmic antibodies

(pANCA) in inflammatory bowel disease (IBD), and to evaluate their discriminative ability

UC and CD (10). The authors concluded that ASCA and pANCA antibodies are specific, but

not sensitive for CD and UC in adult population. S

Similarly, the additional diagnostic value of other serum biomarkers such as

antibodies against exocrine pancreas (PABs), anti-granulocyte macrophage colony-

stimulating factor (anti-GM-CSF) antibodies, or anti-microbial antibodies is small (11).

Likewise, genetic testing does not allow diagnosis of IBD, but further research could evaluate

the potential risk stratification of CD or UC based on genetic markers (12).

Statement 4: Fecal calprotectin is a non-invasive disease activity measure that may be used

to screen for IBD, assess treatment response, evaluate symptoms, and predict recurrence.

Vote: 100% strongly agreed; high-quality evidence

Fecal calprotectin (FC), a neutrophil-derived protein, appears to be the most sensitive

marker of intestinal inflammation in IBD. Calprotectin FC values correlate well with

endoscopic indices of disease activity, and thus allow serial monitoring of disease activity

6
and of treatment success, and can even serve in predicting clinical recurrence or sustained

remission (13-16).

Statement 5: Ileo-colonoscopy with biopsies from inflamed and non-inflamed segments is

necessary for suspected IBD. If the patient has severe acute colitis, sigmoidoscopy is

preferred.

Vote: 100% strongly agreed; high-quality evidence

Ileo-colonoscopy is considered the gold standard investigation when it comes to

diseases of the large bowel disease, as itnd allows direct mucosal visualization, biopsy, and

therapeutic intervention of the colon and terminal ileum. During ileocolonoscopy, tTwo

biopsies should be taken through ileo-colonoscopy from the inflamed areas in order to

diagnose UC and CD. Additional biopsies from the uninflamed areas, and every segment of

the colon, including that of the rectum, especially for the diagnosis of UC, could be used to

help in the diagnosis process.

Infectious colitis is distinguished from IBD by the presence of intact crypt

morphology and acute inflammatory process (17). In this case, biopsies of inflamed and

uninflamed regions are required. Sigmoidoscopy with biopsies may offer a suitable

alternative in patients with severe UC due to the high risk of perforation (18). Later on, after

anfollowing improvement in the condition, ileo-colonoscopy would still be required to

determine disease extension, degree of inflammation, and to rule out CD (18).

Statement 6: There are no endoscopic features unique to CD or UC. The most important

endoscopic features of Ulcerative colitisUC are confluent and continuous colonic

involvement with clear demarcation of inflammation and rectal involvement. The most

crucial endoscopic features of Chron's diseaseCD include perianal involvement, presence of

fistulae, strictures, and discontinuous lesions.

Vote: 90% strongly agreed; high-quality evidence

7
The colonoscopy findings usually show UC as a continuously inflamed segment

involving the distal rectum and extending proximally, loss of vascular markings, granularity,

and friability of the mucosa., as well as dDeep ulcers and bleeding are associated with more

severe cases (19). Colonoscopy features suggestive of CD include mucosal nodularity,

ulcerations (both aphthous and linear), antral thickening, presence of fistulae, and strictures

(20).

Statement 7: Features of chronicity on histological examination of the mucosa such as crypt

architectural distortion, and chronic inflammation are necessary to make the diagnosis of

IBD. Histological findings may help differentiate UC from CD.

Vote: 100% strongly agreed; high-quality evidence

Histological examination of the mucosa plays an important role during the diagnosis

of IBD. For example, iInfectious colitis is distinguished from IBD by the presence of intact

crypt morphology and acute inflammatory process whereas features of chronicity such as

crypt architectural distortion, and chronic inflammation are necessary to make the diagnosis

of IBD (17). Histological features can also aid in differentiating between UC and CD through

the presence of patchy disease, and granulomas which are suggestive of CD rather than UC

(21). However, in UC patients, basal plasmacytosis has been identified as the characteristic

histological feature with the best predictive value for UC diagnosis (22). At least one month

after presentation, generalized mucosal or crypt architectural deformation, mucosal atrophy,

and villous mucosal surface develop (17, 21).

Statement 8: We recommend using cross-sectional imaging modalities, which include,

Magnetic Resonance (MR), Computed Tomography (CT), and small bowel ultrasonography

(SBUS) when radiological evaluation of the small bowel is required for IBD patients.

Vote: 90% strongly agreed; high-quality evidence

8
To assess the small bowel, either magnetic resonance enterography (MRE) or

computed tomography enterography (CTE) should be performed. Both of these studies

require the patient to drink a large volume of neutral contrast, which is used to help highlight

inflammation, strictures, ulcers, and cobble stoning of the intestinal wall (23). Cross-sectional

imaging can also identify extraluminal findings such as fistulae and mesenteric thickening.

A recent meta-analysis showed that the sensitivity and specificity for CTE were 87%

(95% confidence interval [CI], 78%–92%) and 91% (95% CI, 84%–95%), respectively, for

CTE, and 86% (95% CI, 79%–91%) and 93% (95% CI, 84%–97%), respectively, for MRE

(24).

CTE involves administration of ionizing radiation to patients, which is important

because disease monitoring usually occurs on a lifelong basis for patients with IBD and the

cumulative dose of radiation can be significant across decades. In contrast, MRE often

requires the patient to stay in a small, enclosed space, which cand induce claustrophobia.

MRI is also the best current technique to evaluate perianal disease (25), with a 97%

sensitivity, and 96% specificity for the diagnosis of anal fistulae in CD (26).

Computed Tomography (CT) has advantages that include lower cost, less procedure

time, more suitable procedure, widespread availability, less need for anesthesia, and it is

more suitable for patients with contraindications for MRI. Additionally, CT scanning is more

sensitive in determining the presence of abscesses (27).

Ultrasonography also avoids the use of ionizing radiation in the evaluation of the

bowel, and is primarily used to diagnose bowel wall thickening or differential diagnosis

between inflammatory and fibrotic strictures. Ultrasonography has shown a sensitivity of

89% and a specificity of 94.3% in the assessment of patients with known CD, but is less

accurate in detecting proximal lesions (28).

9
Ulcerative colitis Diagnosis

Statement 9: Patients with persistent or recurrent hematochezia and urgency should be

suspected of having UC after excluding infectious etiology.

Vote: 100% strongly agreed; high-quality evidence

More than 90% of patients with active UC report having rectal bleeding. Associated

symptoms generally reflect the severity of mucosal disease, and may differ according to

disease extent (29). L

Loose stools for more than 6 weeks differentiates extensive UC from most cases of

infectious diarrhea (30). Patients with active disease also complain of rectal urgency and

tenesmus. Therefore, these persistent or recurrent symptoms should raise suspicion of

ulcerative colitisUC.

Statement 10: In individuals suspected of having UC, stool testing is recommended to rule

out enteric infection including special testing for Clostridium difficile (C. diff) infection.

Vote: 100% strongly agreed; high-quality evidence

Stool specimens should be obtained to exclude common pathogens and specifically

assayed for Clostridium difficile (C. diff) because the presence of an infection with this

pathogen is a major risk factor for complications, hospitalization, and mortality (31).

Moreover, numerous studies have outlined the link between C. diff Clostridium difficile

infection and IBD relapse (32-34).

Testing is commonly done by PCR or ELISA (Enzyme-linked immunosorbent assay)

(35). C. diff C. difficile isolates should be characterized based on their toxin profile and

antimicrobial resistance pattern (36).

10
Statement 11: If UC is identified by sigmoidoscopy, a future complete ileo-colonoscopy is

recommended to determine the extent and severity of inflammation and to diagnose rule out

CD.

Vote: 80% strongly agreed; high-quality evidence

While UC is often initially diagnosed at flexible (or rigid) sigmoidoscopy, it is important

to confirm the diagnosis, extent and severity of disease by means of full ileo-colonoscopy,

usually within the first year, as this can differentiate between UC and Crohn’s diseaseCD,

provide data for further prediction of the disease’s outcome and risk stratification for

dysplasia, as and influence the patient’s management (37).

Crohn’s disease Diagnosis

Statement 12: In Saudi Arabia, tThe differential diagnosis of intestinal tuberculosis (ITB)

should be considered for individuals with suspected ileocecal CD.

Vote: 80% strongly agreed; high-quality evidence

Patients with ileocecal CD can be misdiagnosed with ITB due to similarities in the

presentation. A comprehensive assessment (history, physical examination, laboratory testing,

endoscopy, histology and radiological examinations) is needed to be able to make the

distinction. Night sweats,sweats, concurrent pulmonary tuberculosis, a positive tuberculin

skin test, antibodies to TBpositive interferon gamma release assay (IGRA) for TB, abdominal

lymphadenopathy, ascites, transverse appearing ulcers, and a patulous ileocecal valve are all

signs suggestive of intestinal tuberculosis (38).

Previous research suggested that ITB lesions were both inflammatory and

proliferative, but in CD, the entire thickness of the intestinal wall was implicated in both

11
inflammation and ulceration (39). CD lesions were wider, and rectal lesions were frequent,

compared tobut ITB lesions that were limited to the right colon (39).

In ITB, necrotic lymph node and contiguous ileocecal involvement are common. A

meta-analysis conducted by Du et al. comprising 692 patients showed that the most reliable

histological characteristics in distinguishing ITB from CD were caseating necrosis, confluent

granulomas, as well as ulcers bordered by epithelioid histiocytes (40).

There is no available validated method that can accurately differentiate between CD

and ITB,ITB; however, several key elements found on colonoscopy, serologyical, and

radiological examinations can help the differential diagnosis between the two entities.

The colonoscopy parameters that are considered supportive of a diagnosis of CD

include: anorectal lesions, longitudinal ulcers, aphthous ulcers, and cobblestone appearance

(41). Patients with intestinal TB usually have involvement of fewer than four segments of the

colon, patulous ileocecal valve, transverse ulcers, and scars or post-inflammatory polyps (41).

Additionally, a positive ASCA serology and proximal small bowel disease may

indicate CD, while a positive quantiferon IGRA test and typical pulmonary lesions could

point out to TB (42). TB PCR performed on intestinal biopsies may aid in the distinction

between the two diseases (43).

Statement 13: Adult CD patients with upper GI symptoms, in contrast to asymptomatic

newly diagnosed patients, should undergo upper GI endoscopy.

Vote: 100% strongly agreed; moderate-quality evidence

Patients with IBD who have upper gastrointestinal symptoms such as nausea,

dyspepsia, and vomiting will benefit from upper GI endoscopy (44). The debate about the use

of esophago-gastroduodenoscopy (EGD) for asymptomatic patients with CD is still

continuing, especially due to recent evidence that suggests a higher prevalence of upper GI

involvement in asymptomatic CD (44). However, when the histological diagnosis of CD from

12
intestinal biopsies is difficult, upper GI endoscopy with evidence of focal gastritis from

histopathological examination may support the diagnosis (45).

Statement 14: If ileo-colonoscopy is normal, in a patient suspected of having CD, EGD and

cross-sectional imaging of the abdomen are recommended.

Vote: 80% strongly agreed; high-quality evidence

As previously mentioned, upper GI endoscopy with evidence of focal gastritis from

histopathological examination may support the diagnosis of CD (45). Moreover, the use of

cross-sectional imaging can further delineate the diagnosis of CD.

Statement 15: Small intestine video capsule endoscopy (VCE) is an alternative choice to

cross-sectional imaging for patients with a clinical apprehension of CD and a normal ileo-

colonoscopy.

Vote: 100% strongly agreed; moderate-quality evidence

Small intestine video capsule endoscopy is a sensitive tool to detect mucosal

abnormalities in the small bowel. The diagnostic capabilities of VCE appear to be superior to

MRE or small intestine contrast ultrasound (SICUS) when evaluating the proximal small

bowel , and radiological findings could have a prognostic value (46).

Statement 16: We recommend assessing the risk of retention prior to using VCE when

stenotic disease is suspected in CD.

Vote: 100% strongly agreed; moderate-quality evidence

Contraindications for VCE include: gastrointestinal obstruction, strictures, and

swallowing disorders (47, 48). Data on retention rates in patients with CD varies from 2% to

13% in patients with established CD, to approximately 1.5% in patients with suspected CD

(49). If small bowel stenosis is not firmly excluded, a patency capsule can be used to confirm

small bowel patency before performing VCE.

13
Statement 17: The diagnosis of CD should be suspected if three or more ulcers were found

in the small intestine after excluding the use of non-steroidal anti-inflammatory drugs within

a month of testing.

Vote: 100% strongly agreed; moderate-quality evidence

Limited data exists regarding the number of ulcerations found during enteroscopy that

are suggestive of CD. A study by Mow et al., reported that multiple ulcerations (≥3

ulcerations) found using wireless capsule enteroscopy were considered diagnostic for CD,

and were observed in 26% of cases (50).

Statement 18: Device-assisted enteroscopy may be used to verify the diagnosis of CD in

patients with negative upper and lower endoscopy and features suspicious of CD on MRI or

small bowel VCE.

Vote: 100% strongly agreed; moderate-quality evidence

Monteiro et al., outlined that 25% of patients with unclassified IBD were found to

have small bowel involvement consistent with CD on VCE examination, but approximately

one third (37%) of them remained unclassified during further follow-up (51). This data

supports the need for further assessment of device-assisted enteroscopy as a diagnostic tool

for CD.

Statement 19: CD should be suspected in patients who have recurrent perianal abscesses or

complicated fistulae.

Vote: 100% strongly agreed; high-quality evidence

Perianal manifestations of CD are represented by fistulae and abscesses, being more

frequent in patients with isolated colonic involvement (52). Clinical and imaging findings are

important for the diagnosis and characterization of perianal disease.

From this point of view, there are two types of fistulae: simple, if they are superficial,

have a single external opening, and lack evidence of complications, or complex, if they are

14
high, have multiple external openings, and are associated with abscesses, rectovaginal

fistulae, or anorectal strictures, rectovaginal fistulas, or anorectal strictures (53).

Proctosigmoidoscopy or Iileo-colonoscopy should be routinely performed in all

patients with perianal CD to assess disease extent, severity, presence of internal openings,

and complications such as strictures and cancer (54).

Classification of inflammatory bowel disease

Statement 20: We recommend using the Montreal classification for adults and the Paris

classification for pediatric patients to describe the disease’s phenotype in patients with UC.

Vote: 100% strongly agreed; high-quality evidence

The Montreal classification was introduced as an attempt to describe the extent and

behavior of CD in more detail, and includes a classification system for UC (55). Since its

introduction, several studies have assessed the inter-observer reliability and validity of the

Montreal classification, and concluded that the inter-observer agreement was good for disease

location, but only moderate/fair for upper gastrointestinal involvement. Moreover, the

Montreal didn’t appear to be a reliable classification system for disease severity in ulcerative

colitisUC (56).

Table I: Montreal classifications of UC (55)

E1 Ulcerative proctitis
E2 Left-sided UC (distal to splenic flexure)
E3 Extensive (proximal to

Extent splenic flexure)


S0 Clinical remission
S1 Mild UC

S2 Moderate UC

15
S3 Severe UC
Severity

The Paris classification of UC (Table II) evaluates the disease’s extent that is

classified into E1, E2, E3 and E4. In E1, ulcerative proctitis, the inflammation is confined to

the rectum. In E2, the inflammation involves a portion of the colorectum that is distal to the

splenic flexure. While in E3, inflammation extends distal to the hepatic flexure, and in E4 the

inflammation extends proximal to hepatic flexure (57). Disease extent should be confirmed

by mapping biopsies, as endoscopic appearance may undervalue the true extent. Determining

disease extent is critical for prognosis, and the risk for undergoing colectomy. Disease

extension is dynamic, and it may progress or regress with time. In the Paris classification of

UC (table I), the severity of the disease is only classified to either S0 (never severe) or S1

(ever severe); the disease is considered severe when the Pediatric Ulcerative Colitis Activity

Index (PUCAI) is elevated to 65 or higher (58).

Table II: Paris classifications of UC (57)

Extent* E1 Ulcerative proctitis


E2 Left-sided UC (distal to splenic flexure)
E3 Extensive (hepatic flexure distally)
Pancolitis (proximal to hepatic flexure)
Severity S0 Never severe†
S1 Ever severe†

*Extent defined as maximal macroscopic inflammation.

Statement 21: We recommend using the Harvey Bradshaw index (HBI) to assess for and

monitor clinical disease activity in CD.

Vote: 90% strongly agreed; high-quality evidence

16
Vote: 90% strongly agreed; high-quality evidence

The Harvey-Bradshaw Index (HBI) is used to assess it’s the endoscopicclinical

disease activity in ileocolonic Crohn's diseaseCD (59, 60). A HBI score less than 5 is defined

as clinical remission, HBI between 5 and 7 as mild disease, HBI between 8 and 16 as

moderate disease, and HBI > 16 as severe disease (60).

Statement 22: We recommend using the Simple Endoscopic Score for Crohn’s Disease

(SES-CD) for assessment of CD endoscopic activity and response to therapy.

Vote: 100% strongly agreed; high-quality evidence

SES-CD score is based on the evaluation of five defined bowel segments (rectum,

sigma + descending colon, transverse colon, ascending colon, and terminal ileum), and in

these segments the presence and size of ulcerations and the extent of the inflammatory area

and stenosis are assessed, then classified in severity as a score of 0–3. The scores for each

individual segment are added together as a sum score (Table III) (61).

Table III: Simple Endoscopic Score for Crohn’s Disease (SES-CD) (61)

Severity 0 1 2 3
Ulcerations none aphtoidAphthous Large ulcers Very large ulcers

ulcers (0.,5 – 2 cm) (>2cm)

(<0.,5 cm)
Ulcerated none <10% 10 – 30% >30%

surface
Inflamed none <50% 50 – 75% >75%

surface
Stenosis none singleSingle, multipleMultiple, notNot passable

passable passable

Composite clinical and endoscopic disease activity in UC

17
Statement 23: In clinical practice, we recommend using the Mayo score for UC as a

composite evaluate tool for disease activity.

Vote: 100% strongly agreed; high-quality evidence

The Mayo Score is a hybrid between clinical and endoscopic variables; stool

frequency, bleeding, inflammatory activity on sigmoidoscopy, overall physician assessment

and daily activities of the patient are assessed (Table IV) (62). In studies, a decrease of the

score by 3 or more is usually taken as therapeutic success. For the assessment of endoscopic

mucosal response, the endoscopic subscore is most often used, and mucosal healing is

diagnosed with an endoscopic subscore of 0 or 1 (but subscore 1 can mean clearly visible

remaining inflammatory activity).

Table IV. Mayo Score (62)

0 1 2 3
Stool frequency 0 1–2 3–4 >5

(aboveAbove-

average)
Bleeding noneNone mild moderate severe
Sigmoidoscopy inactiveInactiv mild moderate severe

e
Overall physician Nnormal mild moderate severe

assessment
Daily activities normalNormal slightly significantly massively

restricted restricted restricted

Endoscopic disease activity in UC

Statement 25: We recommend using the Pediatric Ulcerative Colitis Activity Index (PUCAI)

to evaluate the disease severity and to monitor treatment in children with UC.

Vote: 70% strongly agreed; high-quality evidence

18
The PUCAI is a validated, objective measure of clinical outcome developed to

standardize the reporting of UC disease activity in the pediatric population (Table V) (63).

The PUCAI score has been primarily used within the gastroenterology literature to

characterize disease activity and the effects of medical treatment (64, 65). It is based on 6

quantifiable items easily obtained from a patient’s history, without need for a subjective

abdominal exam, invasive blood testing, or endoscopy (63).

Table V. Pediatric Ulcerative Colitis Activity Index (PUCAI)(63)

Item Points
Abdominal pain

No pain 0

Pain can be ignored 5

Pain cannot be ignored 10

Rectal bleeding

None 0

Small amount only, in less than 50% of 10

stools 20

Small amount in more than 50% of stools 30

Large amount, (N 50% of stool content)

Stool consistency of most stools 0

Formed 5

Partially formed 10

Completely unformed

Number of stools per 24 hours 0

0–2 5

19
3–5 10

6–8 15

>8

Nocturnal stools (any episode causing

wakening 0

No 10

Yes

Activity level 0

No limitation of activity 5

Occasional limitation of activity 10

Severe restricted activity

The Pediatric Crohn’s Disease Activity Index (PDCAI)

Statement 26: We recommend using the Pediatric Crohn’s Disease Activity Index (PDCAI)

to assess the disease severity and to monitor treatment in children with CD.

Vote: 100% strongly agreed; high-quality evidence

The PCDAI focuses on: (1) Subjective reporting of the degree of abdominal pain,

stool pattern, and general well-being; (2) Extra-intestinal manifestations, such as fever,

arthritis, rash, and uveitis; (3) Physical examination findings including abdominal pain,

perirectal disease, extraintestinal manifestations, weight and height; and (4) Laboratory data,

including hematocrit, erythrocyte sedimentation rate (ESR), and serum albumin (66).

This index has high inter-rater reliability, and good construct validity with physician

global assessments of disease activity. Validity has been further demonstrated in several

studies assessing its psychometric properties (67-69).

20
Treatment Goals

Statement 27: In order to evaluate the response to therapy in active UC, a combination of

clinical, endoscopic, and laboratory parameters should be considered.

Vote: 100% strongly agreed; high-quality evidence

The ultimate target of medical therapy is variable among countries because there is no

fully agreed on or validated definition of remission, although many parameters have been

suggested both clinically and endoscopically (70, 71).

Using mucosal healingMH as a treatment target is contentious because of the

implications for clinical practice, with the need for more endoscopic assessment and likely

escalation of therapy in asymptomatic patients (72).

There is lack of clear evidence about the importance of histological remission as well

as endoscopic remission (deep remission) (73, 74). Recent studies suggest that the presence

of endoscopic and histological inflammation is predictive of future flares, lack of sustained

remission, need for corticosteroids, and colectomy (75, 76).

There is a growing consensus among specialists that the target for UC should be

clinical and/or patient-reported remission (defined as absence of rectal bleeding and return to

normal bowel habit) combined with endoscopic remission (Mayo endoscopic subscore of ≤1)

(77).

Recent recommendations from International Organization for the Study of

Inflammatory Bowel Diseases (IOIBD) indicate as a short-term treatment goal the

normalization of CRP (to values under the upper limit of normal), and fecal calprotectinFC

(to 100–250 μg/g), and the change of treatment plan if the target has not been achieved (78).

Statement 28: We recommend using clinical response as an urgent target of treatment in

children as defined below:

21
a) CD: A reduction in the PCDAI score of at least 12.5 points.

b) UC: A reduction in the PUCAI score of at least 20 points.

Vote: 100% strongly agreed; high-quality evidence

Hyams et al., aimed to evaluate the responsiveness of the PCDAI to changes in the

status of patients after therapeutic interventions (79). Clinical response was best reflected by

a decrease in PCDAI of >12.5 points, and subgroup analysis for patients with severe disease

at diagnosis showed a higher mean PCDAI decrease (79). In another study, a change of 12.5

points in the PCDAI was shown to clearly represent a change in physician-assessed severity

(80).

As for PUCAI, a score reduction of at least 20 points was the standard target in

multiple clinical trials for a treatment response evaluation (81-83).

Statement 29: We recommend using clinical response as an urgent target of treatment in

adults as defined below:

a) CD: Minimum 50% decrease in the PRO2 score (stool frequency and abdominal pain).

b) UC: Minimum 50% decrease in the PRO2 score (stool frequency and rectal bleeding).

Vote: 100% strongly agreed; high-quality evidence

Due to the strong correlation of PROs with patient well-being, this target should be

assessed early and frequently throughout the disease course. The STRIDE II study reinforces

a drop of minimum 50% in the PRO2 score for both UC and CD, as a treatment target (78).

Statement 30: We recommend using clinical remission as an intermediate target of treatment

in children, which is defined as:

22
a) CD: an alleviation in in the PCDAI score to a score of <7.5 or <10 points excluding

the height item.

b) UC: a reduction in the PUCAI score to a score of less than 10 points.

Vote: 100% strongly agreed; high-quality evidence

The same IOIBD consensus stated that an alleviation in the PCDAI score to a score of

<7.5 or <10 points excluding the height item for CD, and a reduction in the PUCAI score to a

score of less than 10 points for UC, represent definitions for clinical remission (78).

Statement 31: We recommend using clinical remission as an intermediate target of treatment

in adults, which is defined as:

a) CD: PRO2 (stool frequency ≤3 and abdominal pain ≤1 and) or HBI <5.

b) UC: PRO2 (stool frequency=0 and rectal bleeding=0) or a partial Mayo score of <3

and no score >1.

Vote: 100% strongly agreed; high-quality evidence

We agree with the IOIBD consensus regarding the definition of clinical remission for

adults, and its consideration as an intermediate target of treatment (78).

Statement 32: Fecal calprotectin obtained within 12 weeks of starting medication in IBD

patients may be used as an indicator for the biochemical response.

Vote: 100% strongly agreed; moderate-quality evidence

Fecal calprotectinFC predicted long-term clinical outcomes when measured 12 weeks

after initiating medical treatment (84). A meta-analysis indicated that patients with elevated

fecal calprotectinFC had 53% to 83% probability of relapse during the subsequent 2 to 3

months (85).

Other studies support that the level of fecal calprotectinFC at week 12 to 14 following

anti-TNF initiation is predictive of clinical remission (86, 87).

23
Statement 33: Cross-sectional imaging obtained six to nine months from starting the

treatment can be used to evaluate the transmural response to therapy in CD.

Vote: 90% strongly agreed; moderate-quality evidence

Cross-sectional imaging represented by SBUSIUS, CT, or MRI can be used to

evaluate the transmural response to therapy in CD.

In a prospective multicentric longitudinal study of patients with active CD, all patients

underwent a clinical assessment and sonographic examination at baseline, 12 weeks after

treatment initiation, and after 1 year of treatment, and the authors concluded that sonographic

response after 12 weeks of therapy predicts 1-year sonographic response (88).

Another multicentric trial evaluated the role of IUS SBUS for monitoring treatment

response, and the authors found out that almost all sonographic parameters determined during

IUS SBUS (including bowel wall thickness, vascularization parameters, fibro-fatty

proliferation) showed a significant decrease (89).

The value of CT was assessed in a retrospective study on infliximab-treated patients

with CD (90). Poor-to-fair correlation was found between CT enterography features of

response and improved clinical symptoms or endoscopic appearance, and reduction of CRP

(90). The authors also concluded that only the presence of the ‘comb sign’ on the index CT

enterography was predictive of radiological response (90).

Even though CT might be a suitable method to determine disease activity in CD, it

should not usually be used for monitoring disease activity if MRI or IUS is available due to

radiation concerns.

A prospective single-center trial which evaluated patients with CD treated with anti-

TNF (infliximab or adalimumab) indicated that the Magnetic Resonance Index Of Activity

(MaRIA) score significantly changed at Week 26, and that the overall MaRIA score

correlated well with endoscopic score both at baseline and at Week 26 (91). The authors

24
concluded that the MaRIA has a high accuracy for prediction of endoscopic mucosal healing,

and is a reliable indicator to monitor the use of TNF antagonists in patients with CD (91).

Statement 34: Endoscopic reassessment in UC and CD should be done in cases of patients

with relapse, prolonged disease activity, new unexplained symptoms, and before switching of

treatment.

Vote: 90% strongly agreed; moderate-quality evidence

For follow-up of active disease in UC and CD, endoscopy remains the reference

standard, and several studies determined the benefit of mucosal healing (MH) outlined

through endoscopy in patients with UC and CD.

In a recent meta-analysis, patients with MH had a pooled odds ratio (OR) of 4.50 for

achieving long-term (after at least 52 weeks) clinical remission, 4.15 for remaining free of

colectomy, 8.40 for achieving long-term MH, and 9.70 for achieving long-term

corticosteroid-free clinical remission, when compared with patients without MH (92).

In a prospective multicentric cohort study, the authors showed that endoscopy was the

most sensitive method to detect the earliest mucosal changes, and that severe endoscopic

recurrence at 1 year seems to predict a clinical relapse (93).

Statement 35: Endoscopic healing must be measured through be measured by:

(a) CD: no ulceration or an SES-CD score less than 3 points (e.g., SES-CD ulceration sub

score equal zero);

(b) UC: UCEIS score up to one point or Mayo endoscopic subscore equal of zero.

Vote: 100% strongly agreed; high-quality evidence

We agree with the STRIDE II recommendation regarding the measurement of

endoscopic healing (78).

The Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE) initiative of the

International Organization for the Study of Inflammatory Bowel Diseases (IOIBD) has

25
recommended treatment targets in 2015 for adult patients with IBD. STRIDE II

recommendation for endoscopic healing that it is associated with long term outcome and it

reduce risk of bowl damage. EH was the main treatment procedure in STRIDE II initiative

and scored high point in Delphi like process.

Statement 36: Mucosal healing (MH) should be evaluated endoscopically within 3 to 6

months in UC and 6 to 9 months in CD following treatment initiation in individuals with UC

who have clinically responded to medical therapy.

Vote: 90% strongly agreed; high-quality evidence

We agree with the STRIDE II recommendation regarding the evaluation of mucosal

healing (78).

Statement 37: Histologic remission and transmural healing are not recommended treatment

goals in CD or UC. However, in UC, examination of histologic remission using Robarts

histologic index of severity (RHI) or the Nancy index (NI) can be used as an addition to

endoscopic remission to document a deeper degree of healing. In CD, transmural healing (as

measured by CTE, MRE, or SBUS) is also recommended as a supplement to endoscopic

healing.

Vote: 100% strongly agreed; moderate-quality evidence

The Nancy Index (NI) and Robarts Histopathological Index (RHI) are the two scores

that vary in complexities and features they evaluate (94). However, recent evidence suggests

that their ability to classify UC patients accurately and consistently could be improved (95).

We agree with the STRIDE II recommendation whichrecommendation, which states

that histologic remission and transmural healing are not recommended treatment goals in CD

or UC (78).

Statement 38: In UC and CD, the normalized quality of life related to health and absence of

disability are considered as long-term therapeutic goals.

26
Vote: 100% strongly agreed; moderate-quality evidence

We agree with the STRIDE II recommendation regarding the quality of life for

patients with CD or UC as a long-term therapeutic goal (78).

Statement 39: Prior to starting any biologic therapy, including anti-TNF treatment, patients

with IBD must be screened for latent tuberculosis (TB) using chest radiography and a

purified protein derivative (PPD) skin test and/or an interferon-gamma release assay (IGRA).

Vote: 100% strongly agreed; high-quality evidence

Tuberculosis is an absolute contraindication for biologic therapy (96), so which is

why it must be excluded before the treatment’s initiation using a standard diagnosis protocol,

which that includes chest radiography and a purified protein derivative (PPD) skin test and/or

an interferon-gamma release assay (IGRA test).

Statement 40: Biological therapies should be used in conjunction with preventive anti-

tuberculosis therapy, with at least isoniazid for the first six months, in IBD patients with

confirmed latent TB infection.

Vote: 100% strongly agreed; high-quality evidence

If latent TB infection is confirmed, physicians should associate biological therapies

with preventive anti-tuberculosis therapy for the first 6 months (96).

Treatment Algorithms

Statement 41: A comprehensive risk assessment of CD patients at baseline should be

considered, and the preferred treatment strategy should be suggested according to the risk

profile. A “step-up” approach is recommended for patients who do not have high-risk

features. A “top-down” approach is recommended for patients with high-risk features.

Vote: 100% strongly agreed; high-quality evidence

27
The term 'step-up' refers to a sequential treatment strategy that often begins with a less

effective, potentially less toxic treatment strategy, such as aminosalicylates, antibiotics or

budesonide, with escalation to the highly effective, but potentially more toxic treatment

strategiesoptions, such as prednisone, immunosuppressantsmodulators and biological therapy,

in patients who failed each line of therapy (97).

In this strategy, one would avoid overtreating and unnecessary exposure to the risk of

developing adverse events, especially in a subgroup of patients who may do well with the

standard paradigm. For the same reason of toxicity, physicians are often reluctant to advance

therapy resulting in patients not being adequately treated and developing tissue damage with

the prolonged period of uncontrolled inflammation (98).

The iIdeas of using highly effective, but potentially more toxic treatment strategies

early in the course of a chronic illness to prevent disease progression and disability, has

gained popularity for the treatment of patients with CD. Moreover, the “top-down” strategy

appears to have better promising results for the treatment of CD (99).

Statement 42: The conventional “step-up” approach is the core approach for the treatment of

UC, except for patients who present with acute severe ulcerative colitis (ASUC) that require

hospitalization.

Vote: 100% strongly agreed; high-quality evidence

The treatment of acute severe ulcerative colitisASUC is complex, multidisciplinary,

and requires patient’s hospitalization. Recent data favors the “topstep down approach” using

rescue therapy as an initial medical treatment, but its failure mandates surgical intervention

(100-102).

Crohn’s disease

28
Statement 43: We recommend AGAINST using 5-ASA for the maintenance or even

induction of remission in CD.

Vote: 100% strongly agreed; high-quality evidence

The ECCO team performed a meta-analysis of seven eligible RCTs that compared the

use of oral 5-ASA or sulfphasalazine with placebo in patients with active CD (103). Overall,

there was no significant effect for induction of clinical remission, and among the five trials of

5-ASA alone, there was also no benefit over placebo for inducing clinical remission (103).

Statement 44: Oral Budesonide should only be used to induce clinical remission in

individuals with active mild-to-moderate CD confined to the terminal ileum and/or ascending

colon/cecum.

Vote: 100% strongly agreed; high-quality evidence

A Cochrane systematic review and meta-analysis which compared budesonide at a

dose of 9 mg/day with placebo indicated that budesonide was superior to placebo for

inducing clinical response, and clinical remission in patients with mildly active CD in the

small and/or large intestine limited to the ascending colon (104).

In another Cochrane systematic review and meta-analysis that compared budesonide

at a dose of 9 mg/day with mesalazine up to 4.5 g/day, budesonide was not superior to

mesalazine for inducing clinical remission in patients with mildly active CD in the small

and/or large intestine, but clinical response was more frequently seen in patients receiving

budesonide, which exhibited a good safety profile (105).

Statement 45: Systemic corticosteroids can be used for the induction of remission in active,

moderate-to-severe CD.

Vote: 90% strongly agreed; high-quality evidence

A Cochrane systematic review evaluated data from two2 RCTs regarding the efficacy

of systemic corticosteroids (oral prednisolone or oral methylprednisolone) compared with

29
placebo for the treatment of moderately-to-severely active CD. The results indicated that the

clinical response was more common in patients receiving methylprednisolone as compared

with placebo, and corticosteroids were reported to be twice as effective in inducing clinical

remission than placebo (106).

Statement 46: We recommend AGAINST using thiopurines monotherapy for the remission

and induction of moderate to severe CD. We recommend using thiopurines or parenteral

methotrexate as corticosteroid-sparing agents for maintenance of remission in patients who

are corticosteroid-dependent.

Vote: 80% strongly agreed; high-quality evidence

The pooled analysis of several studies has shown no differences for induction of

remission between thiopurines and placebo (107). Since the quality of evidence is low

regarding the use of thiopurines therapy for the induction of remission and induction of

moderate to severe CD (107), we do not recommend it.

Statement 47: We recommend biologic therapy for induction and maintenance and induction

of remission in patients with moderate-to-severe CD who failed conventional therapy.

Vote: 90% strongly agreed; high-quality evidence

Monoclonal antibodies directed against TNF-α are fast-actingfast acting and potent

anti-inflammatory agents. Anti-TNF therapies approved for the treatment of CD include

infliximab, adalimumab, and certolizumab pegol.

Infliximab is a chimeric mouse-human immunoglobulin [Ig] G1 monoclonal antibody

administered intravenously at a dose of 5 mg/kg at 0-, 2-, and 6-weeks during induction, and

every 8 weeks thereafter. Adalimumab is a fully humanized IgG1 monoclonal antibody given

subcutaneously (SC) at a dose of 160 mg, and then 80 mg 2 weeks after induction, followed

by 40 mg SC every 2 weeks. Certolizumab pegol is a PEGylated Fab fragment against TNF-

30
α, self-administered SC at a dose of 400 mg at Weeks 0, 2, and 4, followed by 400 mg every

4 weeks thereafter.

Data on anti-TNF agents versus placebo (infliximab, adalimumab, and certolizumab

pegol) from several meta-analyses of RCTs support their efficacy for induction of clinical

remission and clinical response (108-110).

The choice of anti-TNF agent depends on patient preference, availability, cost, and

accessibility. However, a recent study revealed that infliximab with AZA and adalimumab

monotherapy were superior to certolizumab pegol for induction of remission (111).

Statement 48: We recommend using adalimumab monotherapy rather than in combination

with an immunosuppressant for induction of clinical remission and response in biologic naïve

CD patients.

Vote: 90% strongly agreed; moderate-quality evidence

One study evaluated the use of combination therapy of adalimumab with thiopurine as

compared with adalimumab monotherapy for the induction of clinical remission in patients

naïve to both therapies. The results showed that combination therapy was not superior to

adalimumab monotherapy for inducing clinical remission (112).

Statement 49: We recommend using thiopurines in combination with infliximab for

induction of remission in patients with moderate-to-severe CD who failed conventional

therapy.

Vote: 90% strongly agreed; moderate-quality evidence

The SONIC (Study of Biologic and Immunomodulator Naive Patients in Crohn’s

Disease) study compared the efficacy of infliximab combined with AZA over infliximab

monotherapy in patients naïve to both therapies, who failed to respond to steroids or 5-ASA.

Combination therapy resulted in higher rates of clinical remission at Week 26 as compared

with infliximab monotherapy, and was also more likely to result in mucosal healing (113).

31
Statement 50: For moderate to severe CD patients who failed both anti-TNF therapy and/or

conventional therapy, ustekinumab is proposed to trigger remission.

Vote: 90% strongly agreed; high-quality evidence

Ustekinumab is an IgG1 monoclonal antibody that binds to the p40 subunit shared by

the pro-inflammatory interleukins 12 and 23 (114). In CD, induction should be given IV

using a weight-based regimen of approximately 6 mg/kg (115).

One systematic review and meta-analysis pooled the results from RCTs in which

ustekinumab was compared with placebo for induction of remission in patients with

moderate-to-severe active luminal CD, and the results showed a significant clinical response

rate when using ustekinumab (116).

Statement 51: For moderate to severe CD patients who failed both anti-TNF therapy and/or

conventional therapy, vedolizumab is proposed to inducetrigger remission.

Vote: 100% strongly agreed; high-quality evidence

Vedolizumab is a monoclonal IgG1 antibody that acts by blocking the α4β7 integrin

resulting in gut-selective anti-inflammatory activity (117). It is administered intravenously at

a fixed dose of 300 mg at 0, 2, and 6 weeks for induction, and every 8 weeks thereafter (118).

Several RCTs evaluated the treatment with vedolizumab or placebo, and reported on

induction of clinical response and clinical remission in adult patients with moderate-to-severe

active CD (117, 119, 120). In all these studies, patients treated with vedolizumab had

significantly higher rates of clinical remission and clinical response.

Statement 52: For moderate to severe CD patients who failed anti-TNF therapy, we

recommend using either ustekinumab or vedolizumab.

Vote: 50% strongly agreed; high-quality evidence

One systematic review and meta-analysis performed an indirect comparison of

ustekinumab and vedolizumab for induction of remission in patients with moderate-to-severe

32
active luminal CD who were non-responsive or intolerant to previous anti-TNF agents (121).

The authors reported no significantly different clinical response and clinical remission rates,

although the quality of data was low.

Perianal Fistulizing Disease

Statement 53: Infliximab is recommended as the first choice of biologic to induce and

maintain remission in complex perianal fistulae in CD.

Vote: 100% strongly agreed; high-quality evidence

Infliximab was the first agent shown to be effective in an RCT for inducing closure of

perianal fistulae and for maintaining this response over 1 year (122).

A meta-analysis of the existing data revealed that infliximab was found to be effective

in inducing fistula healing, and in maintaining clinical fistula healing with no significant risk

of serious adverse effects as compared with placebo (107).

In clinical practice, infliximab is often used in combination with immunosuppressants,

antibiotics, and surgical treatment (123).

Statement 54: Adalimumab can be used to induce and maintain remission in complex

perianal fistulae in CD.

Vote: 90% strongly agreed; high-quality evidence

A meta-analysis of the current data showed that adalimumab was superior when

compared to placebo for fistula healing after 56 weeks (107). Moreover, the open-label

CHOICE trial demonstrated that complete fistula healing could be achieved in patients who

initiated adalimumab after infliximab failure (124).

Statement 55: A combined medical and surgical approach is recommended for patients with

CD and complex perianal fistula.

Vote: 100% strongly agreed; high-quality evidence

33
There is insufficient evidence to support a decision for or against the use of

immunomodulators as a treatment for complex perianal fistula in patients with CD (107).

Further research is needed to reduce uncertainty, but it is reasonable to accept a combined

approach of complex perianal fistula in patients with CD.

Statement 56: We recommend AGAINST using antibiotics for the closure of fistulae in CD

patients with complex perianal fistulae.

Vote: 90% strongly agreed; high-quality evidence

Antibiotics are widely used in the treatment of perianal CD, but most published

studies are uncontrolled (125). Despite the lack of evidence to support their role as

monotherapy in closing perianal fistulae, antibiotics remain indicated and recommended to

treat and control perianal sepsis.

Ulcerative Colitis

Statement 57: Oral and/or topical mesalazine derivatives are recommended as first-line

treatment for the induction and maintenance of remission in mild to moderate UC.

Vote: 100% strongly agreed; high-quality evidence

Oral 5-ASA (5-aminosalicylic acid) is the standard therapy for mild to moderately

active UC. Meta-analyses support the efficacy of oral 5-ASA for induction therapy for mild

to moderately active UC (110, 126). Once daily dosing is as effective as divided doses (127).

Doses ≥2 g/day are more effective than dosages <2 g/day for remission (110). The majority of

patients with mild to moderate UC will respond to 2–3 g 5-ASA (depending on formulation

used) and higher doses can be used in those with more severe symptoms or those not

responding initially.

Statement 58: Budesonide MMX or topical and/or oral corticosteroids are recommended for

induction of remission in UC in patients who failed to respond to mesalazine derivatives.

34
Vote: 100% strongly agreed; high-quality evidence

Randomized controlled trials have shown that oral budesonide MMX 9 mg daily is

significantly more effective than placebo and can induce remission in mild to moderate UC,

being as effective as 5-ASA (128-130).

Another placebo-controlled trial of budesonide MMX in mild to moderately active

UC showed significant benefit in clinical, endoscopic and histological remission (131).

Statement 59: Oral Budesonide MMX is recommended over conventional oral

corticosteroids to induce remission in UC. If conventional oral corticosteroids are used, the

patient should be advised about common and serious side effects of corticosteroids.

Vote: 100% strongly agreed; high-quality evidence

Oral corticosteroids are very effective when used as induction agents, mainly to

control symptoms. However, the use of corticosteroids is limited by significant safety

concerns. On the short term, side effects such as increased risk of infection, weight gain,

acne, glucose intolerance, hypertension, glaucoma, and sleep/psychological disturbances

should be considered and relayed to the patient. Furthermore, corticosteroids should never be

used as maintenance agents as its prolonged use is associated with metabolic bone disease,

cataract formation, adrenal insufficiency, risk of opportunistic infections, diabetes mellitus,

and hypertension. Budesonide MMX may be considered as an alternative to conventional

corticosteroids in patients with mild-moderate UC and failure of response to 5-ASA therapy

(132). B

Budesonide has a lower rate of systemic adverse effects than conventional

corticosteroids (33% vs 55%), and is not associated with adrenal suppression or a significant

reduction in bone mineral density (133).

Statement 60: We recommend using thiopurines to maintain remission in patients with UC

who are corticosteroid resistant or corticosteroid-dependent.

35
Vote: 100% strongly agreed; high-quality evidence

Numerous studies confirm a benefit of thiopurines over placebo for the maintenance

of steroid-induced remission in UC (134, 135). A recent Cochrane review included 232

patients from four maintenance studies of azathioprine versus placebo and showed a benefit

of azathioprine over placebo (136).

Statement 61 IT WAS REMOVED!!!

Statement 62: We recommend using vedolizumab over adalimumab to induce remission in

moderate to severe ambulatory adult UC patient naïve to biologic agents.

Vote: 100% strongly agreed; high-quality evidence

The VARSITY trial evaluated patients with moderately to severely active UC who

had failed conventional therapies and were randomized to intravenous vedolizumab 300 mg

at weeks 0, 2 and 6 and then every 8 weeks, versus adalimumab subcutaneously 160 mg at

week 0, 80 mg at week 2, and then 40 mg fortnightly (137). At 52 weeks, the primary end-

point of clinical remission (a complete Mayo score ≤2 with no subscore >1), and mucosal

healing (Mayo endoscopic subscore ≤1) were significantly higher for the vedolizumab group.

These data provide support for vedolizumab as a first-line biologic option for UC

failing conventional therapy.

Statement 63: In patients with moderately active UC who have failed conventional therapy,

treatment with infliximab, adalimumab, vedolizumab, or ustekinumab is recommended.

Vote: 100% strongly agreed; high-quality evidence

All of the above-mentioned agents could be used as a treatment for patients with

moderately active UC who have failed conventional therapy, but the choice of drug should be

determined by clinical factors, patient choice, cost, likely adherence and local infusion

capacity (138).

36
Statement 64: We recommend using ustekinumab for inductionnitiation and maintenance of

remission in patients with moderate to severely active UC.

Vote: 100% strongly agreed; high-quality evidence

The UNIFI trial investigated ustekinumab as an induction and maintenance therapy in

adults with moderate to severely active UC in adults who had failed to respond or were

intolerant to corticosteroids, immunomodulators, anti-TNF therapy (one or more) or

vedolizumab (139). Patients were randomized 1:1:1 to receive a single IV dose of placebo,

130 mg ustekinumab, or approximately 6 mg/kg ustekinumab. Both active treatment groups

had a significant clinical remission at week 8, endoscopic healing, and clinical response.

Statement 65: Tofacitinib is recommended as a second-line treatment in adult outpatients

with moderate to severe UC who failed biologic agents.

Vote: 60% strongly agreed; high-quality evidence

In a recent meta-analysis, that evaluated tofacitinib as a treatment option of moderate

to severely active ulcerative colitisUC among various biologic agents, showed superior

induction rates of clinical remission (140).

Statement: We recommend AGAINST using Tofacitinib for patients who have a history of

thromboembolic disease, cardiovascular disease, or those ≥50 years old with at least one

cardiovascular risk factor because of an increased risk of thromboembolic events.

Recent data from an open-label study of rheumatoid arthritis patients (over 50 years

with at least one cardiovascular risk factor), comparing tofacitinib 5 mg or 10 mg twice daily

with TNF-inhibitor therapy, have shown a five-fold increase in pulmonary embolus for the

group on 10 mg twice daily tofacitinib compared with TNF inhibitor therapy (141).

It is advisable that the high dose should not be used in patients at increased risk of

pulmonary embolus.

37
Statement 66: We recommend AGAINST methotrexate use to initiate or maintain remission

in adults with moderate to severe UC.

Vote: 100% strongly agreed; high-quality evidence

A Cochrane review in 2015 of methotrexate use in comparison to placebo, 5-ASA,

sulfasalazine and mercaptopurine does not support the role of Methotrexate in the

maintenance of remission in UC (142). Moreover, a European double-blind randomized trial

evaluated patients who were allocated to 25 mg/week parenteral methotrexate versus placebo

alongside prednisolone for a flare of UC, and the results showed that Methotrexate was not

superior to placebo for the primary end-point of steroid-free remission at week 16 (Mayo

score ≤2 with no item >1 and complete withdrawal of steroids and no use of another

immunosuppressant (IS), anti-TNF therapy or colectomy) (143).

Statement 67: In moderate to severe UC, we recommend combining infliximab with

thiopurines over Infliximab monotherapy. There is no sufficient evidence to recommend

combining other biological therapies with thiopurines.

Vote: 100% strongly agreed; moderate-quality evidence

In the UC SUCCESS study, patients in whom corticosteroid therapy had failed and

who were receiving infliximab and azathioprine combination therapy had significantly higher

remission rates at week 16, compared with infliximab alone (144). A recent technical review

from AGA supports the association between infliximab and thiopurines for the treatment of

moderate to severe UC (145).

Statement 68: We recommend AGAINST 5-ASA continuation for maintenance of remission

in adult ambulatory patients with moderate to severe UC who have attained remission with

use of immunomodulators, and/or biologic agents or tofacitinib.

Vote: 100% strongly agreed; moderate-quality evidence

38
There is current no strong evidence that supports the use of 5-ASA continuation for

maintenance of remission in adult ambulatory patients with moderate to severe UC who have

attained remission with use of immunomodulators, and/or biologic agents or tofacitinib (146,

147).

39
Biologics
The use of biologics in moderate to severe UC

Statement 69: Biological medications should be selected based on patient preferences,

availability, cost, risk of infection, presence of extra intestinal manifestations of IBD, and the

desired onset of response.

Vote: 70% strongly agreed; high-quality evidence

Several studies have outlined the following factors as selection criteria for a biological

agent selection: patient preferences, availability, cost, risk of infection, presence of extra

intestinal manifestations of IBD, and the desired onset of response (148-150).

Statement 70: We recommend infliximab, adalimumab, vedolizumab or ustekinumab for

patients on high-dose mesalazine maintenance therapy requiring two or more courses of

corticosteroids in the preceding year or who patients developed corticosteroid dependence or

refractory condition.

Vote: 100% strongly agreed; high-quality evidence

In a technical review, data from 16 RCTs comparing the TNFα antagonists,

vedolizumab, tofacitinib and ustekinumab to placebo as treatment options,options was

analyzed. The results showed that all active interventions were superior to placebo for

induction of remission, and for maintenance of remission. Moreover, it was concluded that all

medications were well-toleratedwell tolerated with low rates of serious adverse events (145).

The use of biologics in moderate to severe CD

Statement 71: We recommend using TNF inhibitors (Certolizumab pegol, adalimumab or

infliximab), ustekinumab or vedolizumab, for adult patients with moderate to severely active

CD to induce and sustain remission.

Vote: 100% strongly agreed; high-quality evidence

40
We agree with the current recommendations regarding the possible use of the

following biological agents for induce and maintenance of remission in adult patients with

moderate to severely active CD (107).

Statement 72: We recommend starting TNF inhibitors (infliximab, adalimumab, or

certolizumab pegol), vedolizumab, or ustekinumab for patients with severely active CD who

did not tolerate or had an inadequate response to conventional therapy such as

immunosuppressants or corticosteroids.

Vote: 100% strongly agreed; high-quality evidence

The UNIFI trial investigated ustekinumab as induction and maintenance therapy in

moderate to severely active UC in adults who had failed to respond or were intolerant to

corticosteroids, immunomodulators, anti-TNF therapy (one or more) or vedolizumab (139).

Patients were randomized 1:1:1 to receive a single IV dose of placebo, 130 mg ustekinumab,

or approximately 6 mg/kg ustekinumab. Both active treatment groups had a significant

clinical remission at week 8, endoscopic healing, and clinical response.

The VARSITY trial evaluated patients with moderately to severely active UC who had

failed conventional therapies and were randomized to intravenous vedolizumab 300 mg at

weeks 0, 2 and 6 and then every 8 weeks, versus adalimumab subcutaneously 160 mg at week

0, 80 mg at week 2, and then 40 mg fortnightly (137). At 52 weeks, the primary end-point of

clinical remission, and mucosal healing were significantly higher for the vedolizumab group.

The series of PRECISE studies have shown that Certolizumab pegol offers a rapid

response and sustained response and remission for patients with moderate to severe CD,

regardless of their prior exposure to anti-TNF therapy and concomitant medication use (151-

154).

41
Statement 73: We recommend an early start of biological therapy for patients with CD who

have high-risk features.

Vote: 100% strongly agreed; high-quality evidence

Early initiation of biologic therapy in UC may help prevent disease-related

complications, such as colon cancer, hospitalizations, and surgery (155). The AGA Institute

Ulcerative Colitis Clinical Care Pathway (156), suggested that early therapy with a biologic

agent should be considered in patients who have factors associated with high colectomy risk

or worse prognosis. These factors include extensive colitis, deep ulcers, age <40, elevated C-

reactive protein (CRP) and erythrocyte sedimentation rate (ESR), steroid-requiring disease,

history of hospitalization, Clostridium difficile infection, and cytomegalovirus (CMV)

infection (156). However, more recent guidelines primarily used disease severity to guide

when biologic therapy is used, which was largely defined by the traditional Truelove-Witts

criteria and Mayo score (157).

Statement 74: We recommend combining Infliximab with methotrexate or thiopurine instead

of using infliximab monotherapy for inducing and maintaining remission in active CD

patients.

Vote: 100% strongly agreed; high-quality evidence

The Study of Biologic and Immunomodulator Naïve Patients in CD (SONIC) trial

evaluated patients with moderate-to-severe CD, naïve to both immunomodulators and anti-

TNF-α drugs, who were randomized to receive eitherreceive azathioprine monotherapy,

infliximab monotherapy, or combination therapy with azathioprine and infliximab. The

combined therapy achieved at week 26 a higher rate of corticosteroid-free clinical remission,

mucosal healing, and an overall better outcome (113). In another study by D’Haens et al., the

42
early combined therapy was superior to conventional therapy for the induction of clinical

remission (158).

A 2020 meta-analysis comparatively evaluated the monotherapy with methotrexate

and the combined methotrexate-infliximab therapy as an option for inducing and maintaining

remission in active CD and UC patients (159). The results did not support the use of

combined therapy for induction and maintaining remission in CD and UC patients, although

monotherapy with methotrexate appeared to be effective for maintenance of clinical

remission in CD (159).

Further research will be needed to assess the efficacy of combined methotrexate-

infliximab therapy for the remission induction in CD patients.

Statement 75: We recommend starting vedolizumab for patients ≥65 years old, patients with

history of a recent infection, and for individuals at higher risk of infection or malignancy. We

recommend using vedolizumab for 2 to 30 weeks for patients with moderate to severe

fistulizing CD.

Vote: 100% strongly agreed; high-quality evidence

Vedolizumab can be used as a monotherapy or in combination with an

immunomodulator for the induction of symptomatic remission in patients with moderately to

severely active CD (160).

Several studies have demonstrated significant risks associated with the use of anti-

tumor necrosis factor (anti-TNF) therapies, corticosteroids, and thiopurines for the treatment

of IBD patients such as higher risks of malignancy and opportunistic infections in elderly

patients (161-163). Although the safety profile of vedolizumab is proven by several papers,

limited data exists regarding the safety and efficacy of this drug in older populations (118,

164). However, the patient’s age ≥65 years should not be considered as a contraindication for

vedolizumab administration.

43
A meta-analysis by Bonovas et al., evaluated the risk of infection and malignancy in

adults with IBD treated with various biological agents (165). The authors concluded that

biologic agents increase the risk of opportunistic infections in patients with IBD, but not the

risk of serious infections. Therefore, it is reasonable to accept the use of vedolizumab for

individuals at higher risk of infection or malignancy who are under careful monitorization.

An exploratory analysis of data from GEMINI 2 revealed the efficacy of vedolizumab

treatment for patients with moderately to severely active CD, who had a history of fistulizing

disease or an active draining fistula (166). After 6 weeks of induction treatment with

vedolizumab (intravenous doses of 300 mg at Weeks 0 and 2), the patients were randomized

to receive either placebo or vedolizumab every 8 weeks or every 4 weeks, and were entered

into a 46-week maintenance phase. The authors found out higher rates of fistula closure and

decreased time of recovery for patients treated with vedolizumab (166).

Sequencing of medications

Statement 76: If disease relapse occurs with vedolizumab therapy, dose-escalation (typically

by shortening dosing interval to every four weeks) should be considered while evaluating for

co-existing or triggering factors.

Vote: 90% strongly agreed; high-quality evidence

In a retrospective study, Perry et al., investigated the efficacy of vedolizumab dose-

escalation in a group of patients with UC. Amongst their cohort of 22 patients treated with

vedolizumab with a partial response to standard 8-weekly dosing, almost half were observed

to achieve remission (partial Mayo score of 0 or 1) upon dose-escalation to 4-weekly (167).

These findings are in accordance with a metanalysis that demonstrated a random-

effects pooled efficacy rate of 53.8% (114), and with a small cohort study of 36 dose-

escalated patients with CD or UC, in which a 50% response rate was observed (168).

44
Statement 77: If disease relapse occurs with ustekinumab therapy, dose-escalation (typically

by decreasing the dosing interval to every four weeks) should be considered while evaluating

for co-existing or exacerbating factors.

Vote: 100% strongly agreed; high-quality evidence

Numerous recent studies have investigated the ustekinumab dose-escalation for CD

relapse. In a retrospective study by Ollech et al., shortening the ustekinumab 90 mg dose

interval to 4 weeks for patients with CD who did not respond to doses every 8 weeks

improved clinical and biological indices of disease activity, without severe adverse effects

(169).

These results are in accordance with another retrospective study whichstudy, which

evaluated the ustekinumab dose-escalation strategy for selected CD patients who failed to

achieve remission on standard Q8 week regimen. The authors outlined that the dose-

escalation strategy improved clinical outcomes, prevented worsening disease severity, and

positively impacted CRP and albumin levels (170).

Finally, a multicentric retrospective cohort study assessed the effectiveness of dose-

escalation of ustekinumab, and concluded that intensification of ustekinumab maintenance

dosage was effective in over 50% of the patients (171).

Interventions based on therapeutic drug monitoring for patients with IBD and loss of
response to anti-TNF agents

Statement 78: In patients failing anti-TNF therapy, we recommend optimizing the dose,

switching to another anti-TNF agent, or switching to a different class of such as ustekinumab

or vedolizumab, based on serum drug level (trough) and the presence of anti-drug antibodies

(ADAs).

Vote: 100% strongly agreed; high-quality evidence

45
The majority of patients treated with anti-tumor necrosis factor (TNF) therapy

develop anti-drug antibodies (ADAs), which might result in loss of treatment efficacy.,

Thiswhich usually occurs within 12 months after the onset of treatment (172). Therefore, the

measurement of anti-TNF antibody trough levels (TL), and the determination of ADA

presence are frequently performed to optimize the management of patients with IBD (173).

The current guidelines recommend optimizing the dose or switching to another anti-

TNF agent in patients failing anti-TNF therapy (4).

Ustekinumab and vedolizumab are an option for patients who have not achieved

adequate response with anti-TNF agents despite adequate drug concentrations, and can also

be used as potential first-line treatments (174).

In the GEMINI 1 trial of vedolizumab, over 40% of patients with UC were prior TNF

failures. In this study, the response rates at week 6 for vedolizumab vs placebo were 47% vs

25% (118). However, a post-hoc analysis revealed that the rates of response at week 6 were

53% for patients naive to anti-TNF therapy and 39% for patients with prior anti-TNF failure

(175).

In the GEMINI 2 study of vedolizumab in patients with CD, almost half of the cohort

consisted of patients who had previously failed anti-TNF therapy. Week 6 clinical remission

for vedolizumab vs placebo was 14.5% vs 6.8% (117). However, CD patients who had failed

anti-TNF therapy had a rate of remission at week 6 of 15% compared to 12% of patients who

were treated with placebo (120).

The UNITI-1 trial, which evaluated patients with CD and included a large number of

patients with prior anti-TNF failure, had a week 6 response of 34.3% and 33.7% for patients

treated with 130 mg or 6 mg/kg of ustekinumab, respectively, vs 21.5% for the placebo group

(41). In UNITI-2, in which the majority of patients were naive to treatment, response to

46
treatment was 52.7% and 55.0% for ustekinumab dosing of 130 mg or 6 mg/kg, respectively,

vs 23.0% for placebo (41).

Statement 79: In patients with low serum drug levels and positive ADAs, we recommend

switching to an alternative anti-TNF agent, especially in the presence of high ADA titers.

Vote: 100% strongly agreed; high-quality evidence

In patients with low titers of ADAs, drug concentrations may remain high enough to

be effective, while in patients who develop high titers of ADAs, a substantial portion of the

drug will be neutralized, and is likely to produce a clinical non-response over time (176).

Several studied support patients the switch to a different anti-TNF agent for patients

who developed high titers of ADAs (177, 178).

A recent meta-analysis evaluated the efficacy and safety of a second anti-TNF agent

(infliximab, adalimumab, or certolizumab-pegol) after primary/secondary failure or

intolerance to a first drug. The efficacy of a second anti-TNF in CD patients largely depends

on the cause for switching. The authors concluded that the remission rate is higher when the

reason to withdraw the first anti-TNF is intolerance (61%), compared with secondary (45%)

or primary failure (30%) (179).

Statement 80: Patients with low serum drug levels and negative ADAs require dose

optimization by either dose-escalation or shortening of the dosing interval.

Vote: 100% strongly agreed; high-quality evidence

Studies suggest that a low level of drug antibodies, can be overcome by dose-

escalation of anti-TNF therapy or addition of an immunomodulator. In a retrospective case

series of twelve IBD patients who were on infliximab or adalimumab therapy, and were

found to have detectable but low-level antidrug antibodies, dose-escalation of the drug

resulted in a resolution of antidrug antibodies, and improved disease activity (180).

47
In a recent observational study, the authors evaluated different predictive external

models in order to help individualize infliximab dosinginfliximab-dosing regimens. The

authors identified two models with the highest classification accuracy which indicated dose-

escalations (for trough concentrations < 5 µg/mL) in 88% of cases, thus questioning

population pharmacokinetic modeling as a way to individualize infliximab dosing (181).

Statement 81: Patients who do not respond to anti TNF therapy while having with adequate

serum drug levels and negative ADAs (primary non response (PNR)) require switching

outside the anti-TNF class to another biological agent.

Vote: 100% strongly agreed; high-quality evidence

For patients with primary non-response PNR to one anti-TNF, the likelihood that they

will respond to a second is small, and switching to a different class of drugs appears to be

more appropriate. Recent data suggests that drug levels in primary non-respondersPNRs are

often lower than in responders, and antibody formation can be a significant factor within a

few weeks of treatment initiation (182).

48
Chapter 2. Special Patient population
Using of biologics in pregnancy

Statement 82: We recommend that individuals females with active disease or who are high-

risk of disease recurrence to continue their medications during pregnancy. In contrast, those

with inactive disease who choose to cease medication can do so at the beginning of the 3rd

trimester.

Vote: 100% strongly agreed; high-quality evidence

Women with an active IBD prior to conception have increased likelihood of adverse

pregnancy outcomes such as premature birth, intrauterine growth restriction, and spontaneous

abortion (183-185). Therefore, it is reasonable to recommend continuation of their

medication during pregnancy.

Discontinuing anti-TNF therapy during pregnancy, in order to minimize exposure to

the fetus, is accepted if there is no significant increase in the risk of disease flare. Several

49
studies on pregnant women with quiescent IBD did not show any increase in risk of flare if

anti-TNF therapy is stopped between 25 and 30 weeks of gestation(186, 187).

Statement 83: For infants who have been exposed to biologic treatments in utero, we

recommend delaying BCG immunization until at least six months after birth and not giving

the Rotavirus vaccine. Non-live vaccines can be administered in accordance with

conventional immunization protocols.

Vote: 100% strongly agreed; high-quality evidence

Several studies support the use of inactivated vaccines for the immunization of

newborns who were exposed to anti-TNF drugs, vedolizumab and ustekinumab in utero (188,

189).

On the other hand, current data suggests that the BCG vaccine should be deferred to 6

months of age, and rotavirus vaccine should not be given at all (as there is no value in giving

rotavirus later than 6 months) (190-192).

Statement 84: Throughout pregnancy, we recommend using phthalate-free 5-ASA,

thiopurines, and anti-TNF to maintain remission. Corticosteroids should be reserved for the

treatment of flares.

Vote: 100% strongly agreed; high-quality evidence

Over the last two decades, both topical 5-ASAs and non-enteric coated formulations

preparations based on 5-ASA have remained a standard of IBD therapy (193, 194). In a

cohort study, Bell et al., (195) evaluated pregnant patients with distal colitis on maintenance

with topical 5-ASA therapies at the time of conception, and found that 5-ASA was safe and

effective for managing distal colitis during pregnancy. Marteau et al., also conducted a study

in which pregnant IBD patients were monitored while taking between 1-4 g/d of mesalamine

microgranules, and found no serious complications during the course of pregnancy, nor did

they find any adverse fetal outcome (196).

50
AZA and 6-MP are pregnancy category “D” drugs, and because of their cytotoxicity,

and potential risk of birth defects, they should be used with great care during pregnancy

(197).

TNF-α inhibitors are currently the most commonly used drugs for the treatment of

IBD in pregnancy, and are considered pregnancy category “B” drugs (188). Biologics like

infliximab and adalimumab cross the placenta, especially during the third trimester (188).

Mahadevan et al., evaluated pregnant patients with IBD being treated with infliximab,

adalimumab, or certolizumab-pegol, and compared concentrations of these biologics in infant

and cord blood with concentrations in the mother’s circulation (198). The levels of infliximab

and adalimumab were elevated in infant and cord blood compared to their respective

maternal levels, with the median level of infliximab in cord blood being 60% higher than that

of the mother.

Statement 85: We recommend using mainly MRI or ultrasound for imaging when required

during pregnancy to minimize radiation exposure. If required, endoscopy can also be

performed, ideally in the second trimester.

Vote: 90% strongly agreed; high-quality evidence

The main advantage of MRI is the ability to image deep soft tissue structures in a

manner that is less operator- dependent and does not use utilize ionizing radiation, thus

making it ideal for evaluation of pregnant patients with IBD.

Endoscopy is the most definitive method of monitoring and evaluating IBD’s activity.

However, endoscopic procedures have been theorized to pose a threat to the fetus through the

possibility of intra-procedural maternal hypoxia and hypotension, which can cause fetal

hypoxia and potential demise, thus being preferred in the second trimester if necessary (199).

Statement 86: Venous thromboembolism prevention is crucial for hospitalized patients,

especially following C-section, or if the patient is unwell.

51
Vote: 100% strongly agreed; high-quality evidence

Venous thromboembolism prophylaxis is mandatory after cesarean section, and for

pregnant patients with reduced mobility, with Fondaparinux being the most utilized

anticoagulant worldwide (200).

Statement 87: Engaging the multidisciplinary team and the patient in your decision-making

process during pregnancy is recommended, and should include an obstetrician with relevant

experience.

Vote: 100% strongly agreed; high-quality evidence

A multidisciplinary team (MDT) is the best approach for an individualized management

of patients diagnosed with IBD. During pregnancy, the presence of aAn experienced

obstetrician in the MDT is helpful in monitoring the mother’s and fetal wellbeing, and can

decide the appropriate mode of delivery of the fetus (201).

Statement 88: Except for active perianal disease as well as an ileocecal anastomosis or

ileoanal pouch (where a CS is usually favored), the delivery mode must be determined by

patient’s preference and obstetric factors.

Vote: 100% strongly agreed; moderate-quality evidence

Perianal disease and ileo-cecal anastomosis are important factors whichfactors, which

can obstruct the mechanism of labourlabor. Therefore,, in these circumstances are impose

delivery through cesarean section rather than vaginally is preferred. Otherwise, the mode of

delivery is dictated by obstetrical factors such as acute fetal distress, fetal dystocia, placental

abnormalities, etc (201).

Statement 89: It is recommended to continue using safe medications throughout pregnancy

and breastfeeding periods.

Vote: 100% strongly agreed; moderate-quality evidence

52
Although not without risk of adverse effects, 5-ASA derivaivatives, thiopurines, and anti-

TNF agents can be used for the treatment of IBD during pregnancy and breastfeeding, as long

as the mother is correctly supervised by an experienced obstetrician (199).

Statement 90: We recommend breastfeeding as a source of nutrition for infants of mothers

with IBD.

Vote: 100% strongly agreed; high-quality evidence

Breastfed infants of mothers receiving biologics, immunosuppressants or combination

therapy have similar risks of infection, and similar milestone achievement at 12 months to

non-breastfed infants, even though low levels of infliximab, adalimumab, certolizumab,

natalizumab and ustekinumab can be detected in breast milk (202).

Statement 91: We recommend that pregnant women diagnosed with IBD on oral and/or

rectal 5-ASA as a maintenance treatment continue to take it during pregnancy. Phthalate-

containing 5-ASA formulations should be avoided.

Vote: 100% strongly agreed; high-quality evidence

Over the last two decades, both topical 5-ASAs and non-enteric coated formulations

preparations based on 5-ASA have remained a standard of IBD therapy (193, 194), and

phthalate-free 5-ASA drugs should be used during pregnancy and breastfeeding (203).

Statement 92: We recommend that pregnant women diagnosed with IBD on thiopurine as a

maintenance treatment continue to take it during pregnancy.

Vote: 100% strongly agreed; high-quality evidence

During pregnancy, the active metabolite 6-thioguanine (6-TG) crosses the placenta, but

the prodrugs AZA and 6-MP do not (204). Recent studies showed no increased risk of

adverse pregnancy outcome in the case of thiopurine use during pregnancy, accordinglyso

thiopurines cancould be used as a maintenance therapy during pregnancy (205, 206).

53
Statement 93: We recommend continuous usage of anti-TNF maintenance treatment in

pregnant women diagnosed with IBD.

Vote: 100% strongly agreed; high-quality evidence

A systematic review showed that anti-TNF therapy does not increase the risk of

unfavorable pregnancy outcomes among women with IBD (207). The long-term effects of in

utero exposure to anti-TNF are not extensively explored. A recent study shows a normal

health outcome and first year development in children exposed to anti-TNF, comparable to

children of non-IBD controls (208).

Statement 94: We recommend that pregnant women with UC suffering from a mild-to-

moderate flare while on 5-ASA maintenance therapy must modify their combination oral and

rectal 5-ASA medication to achieve symptomatic remission.

Vote: 100% strongly agreed; high-quality evidence

We agree with the British Society of Gastroenterology recommendation regarding the

modification of combination oral and rectal 5-ASA medication to achieve symptomatic

remission for pregnant patients suffering from a mild-to-moderate flare while on 5-ASA

maintenance therapy (209).

Statement 95: We recommend systemic corticosteroids or anti-TNF therapy to achieve

symptomatic remission in pregnant women diagnosed with IBD who experience a disease

flare while on optimum 5-ASA or thiopurine maintenance therapy.

Vote: 90% strongly agreed; high-quality evidence

We agree with general guidelines and support the addition of systemic corticosteroids

or anti-TNF therapy to achieve symptomatic remission in pregnant women diagnosed with

IBD who experience a disease flare while on optimum 5-ASA or thiopurine maintenance

therapy (209).

54
Statement 96: We recommend that methotrexate should be stopped at least three months

before pregnancy and until breastfeeding is stopped to avoid detrimental effects on the infant.

Vote: 100% strongly agreed; high-quality evidence

Methotrexate is contraindicated in pregnancy (category X), because it is an

abortifacient and has teratogenic effects, such as causing the development of craniofacial

abnormalities, limb defects, and CNS defects as anencephaly, hydrocephaly, and

meningomyelopathy, especially with first-trimester exposure (210). Because its active

metabolites have a long half-life, methotrexate must be discontinued at least 3 months before

conception.

Diagnostic workup of children with suspected IBD

Statement 97: Enteric infections should be excluded as a source of symptoms in children

with suspected IBD before endoscopy. Bacterial infections, such as Clostridium difficile,

should be ruled out by a microbiological stool examination.

Vote: 100% strongly agreed; high-quality evidence

Enteric infections represent an important differential diagnosis for IBD in pediatric

population, so that it is mandatory to exclude them before endoscopy (209). Moreover,

Clostridium difficile is more frequently associated with IBD in both adult and pediatric

population with IBD, so it is reasonable to rule out this infection by a microbiological stool

examination (211, 212).

Statement 98: Complete blood count, at least two inflammatory markers (CRP, fecal

calprotectinFC or fecal lactoferrin), albumin, transaminases, and GGT, should all be included

in the initial blood tests. Fecal calprotectinFC correlates with detection of intestinal

inflammation more than any blood markers.

Vote: 100% strongly agreed; high-quality evidence

55
Multiple blood tests may appear to be abnormal in Ipatient with IBD. Examples

include: complete blood cell count (decreased hemoglobin or elevated total white cell count

and platelet count), serum albumin (decreased), and inflammatory markers such as CRP and

ESR, both of which are typically elevated in active disease.

Data from pediatric IBD registries indicate that at the time of diagnosis 54% of

children with mild UC and 21% of children with mild CD have normal results for the

combination of hemoglobin, albumin, CRP, and ESR diagnosis (213). Examination of

transaminases and GGT should be performed to screen for IBD-associated extraintestinal

disease such as hepatobiliary disease (214).

The most important fecal surrogate markers for detection of inflammation at diagnosis

include FC and lactoferrin, both being important tools for identifying the presence of

intestinal inflammation with high sensitivity (215, 216).

In a recent prospective study FC was elevated at diagnosis in 95% of 60 unselected

pediatric patients with CD, whereas only 86% of the patients had an increased CRP and 83%

an elevated ESR (67). The combination of any 2 of these 3 markers appears to possess a

higher sensitivity (217).

FC levels at diagnosis of pediatric IBD are superior to blood markers as a diagnostic

marker for intestinal inflammation, and discriminate IBD from other extraintestinal

inflammatory conditions as well as intestinal noninflammatory conditions. In a recent large

case-control study of FC, area under the receiver operating characteristic curve of FC to

diagnose IBD was 0.93, considerably higher than the AUC of blood inflammatory markers

(218).

Statement 99: All children with suspected IBD should have an ileo-colonoscopy and an

EGD as part of their first evaluation.

Vote: 100% strongly agreed; high-quality evidence

56
Ileo-colonoscopy (and biopsies) is the most essential part of the diagnostic workup in

pediatric IBD. The diagnostic yield of ileo-colonoscopy including histology is reported to be

about 13% in pediatric patients diagnosed with IBD (219).

The European consensus statements on diagnosis and management of UC recommend

that ‘‘multiple’’ biopsies from 5 sites around the colon (including the rectum), and the ileum

should be taken, and a minimum of 2 samples from each of these sites should be obtained for

a reliable diagnosis (185).

In pediatric populations, isolated upper GI CD occurs more commonly than in adults.

Thus, an EGD is recommended as part of the initial evaluation of children with suspected

IBD, regardless of upper GI symptoms (220). Upper endoscopy is also useful in evaluation

for celiac disease which can have a similar presentation to IBD in both the adult and pediatric

populations (221).

Statement 100: Even if there are no macroscopic abnormalities, multiple biopsies (2 or more

per section) should be taken from all parts of the accessible gastrointestinal tract.

Vote: 100% strongly agreed; high-quality evidence

The European consensus statements on diagnosis and management of UC reiterated the

importance of minimum 2 samples obtained through biopsy from 5 sites around the colon,

and the ileum, even though no macroscopic abnormalities are identified on endoscopic

examination (185).

Statement 101: At the time of diagnosis, we recommend using MRE over CT scan and

fluoroscopy as the imaging technique of choice in pediatric IBD because of its excellent

diagnostic accuracy and less radiation exposure.

Vote: 100% strongly agreed; high-quality evidence

MRI is the preferred test for imaging the small bowel at diagnosis because it can

detect changes that are characteristic of IBD, and estimate both the extent of intestinal

57
inflammation and the degree of damage (strictures or penetrating disease). It has the

advantage over CT or fluoroscopy of no irradiation.

A systematic review and meta-analysis of studies about cases of suspected pediatric

IBD has confirmed that MRE is sensitive and specific for diagnosis of PIBD and that it

should supersede conventional fluoroscopy as the small bowel imaging technique in centers

with appropriate expertise (222). The pooled sensitivity and specificity in this study for MRE

detection of active terminal ileal CD of 84% and 97%, respectively.

Pelvic MRI is recommended for the evaluation of patients with CD with suspected or

proven perianal involvement, allowing evaluation of extent and location of perianal fistulas

and abscesses, thus providing critical information for both surgical management and for

assessment of response to medical therapy (223).

Statement 102: VCE is an alternative option for identifying small intestinal mucosal lesions

in pediatrics with suspected CD in whom conventional endoscopy and imaging techniques

have been non-diagnostic, or MRE cannot be done.

Vote: 100% strongly agreed; high-quality evidence

Wireless VCE is the best alternative to MRE for investigating the SB, and it detects

mucosal abnormalities. The main advantages of WCE are the ability to visualize the entire

SB with minimal discomfort, and to detect mucosal lesions with a higher sensitivity than

MRE (224). In a meta-analysis in PIBD, the diagnostic yield for WCE ranged from 58% to

72% (225).

Contraindications include intestinal strictures, previous abdominal surgery (relative),

severe disease with systemic features, and children <1 year (226, 227).

Statement 103: SBUS is a useful screening technique in the initial diagnostic workup of

pediatric patients with suspected IBD, but because of its low sensitivity, it should be

supplemented by more sensitive small bowel imaging.

58
Vote: 100% strongly agreed; moderate-quality evidence

SBUS has the advantages of non-invasiveness, low-cost, and widespread availability,

making it a useful screening technique for IBD (228).

SBUS accurately detects, locates, and characterizes inflammation of the bowel wall

and assesses peri-intestinal abnormalities, with a good negative predictive value for IBD,

higher for CD than for UC (229).

Comparative studies between bowel USSBUS and ileo-colonoscopy and histology in

detecting CD lesions at the terminal ileum have shown an overall sensitivity and specificity

of 74% to 88% and 78% to 93%, respectively (230).

Statement 104: All patients with infantile IBD presenting in the first two years of life should

be evaluated for primary immune deficiency.

Vote: 100% strongly agreed; moderate-quality evidence

It appears that patients with very early onset of IBD, until the first 2 years of life, may

have an underlying genetic basis (231). A large proportion of monogenic etiologies reflect

underlying primary immune deficiencies (PIDs), highlighting the importance of a

dysregulated immune system in very early onset of IBD (232).

The evaluation of primary immune deficiencies in patients with IBD is complex, and

should be performed by a multidisciplinary team. It includes, among the standard evaluation

of IBD in children, genetic testing, analysis of immunoglobulin classes (IgA, IgG, IgM, IgE),

and lymphocyte subsets by flow cytometry (233).

Statement 105: We recommend using the PUCAI in children with UC to monitor disease

activity at each visit, and to consider reviewing therapy whenever the PUCAI ≥ 10 points.

Vote: 100% strongly agreed; high-quality evidence

59
We agree with the Guideline from the European Crohn’s and Colitis Organization and

European Society of Pediatric Gastroenterology, Hepatology and Nutrition, that recommends

using the PUCAI in children with UC to monitor disease activity at each visit, and to consider

reviewing therapy whenever the PUCAI ≥ 10 points (234).

Statement 106: A ileo-colonoscopic examination is suggested at diagnosis, before

substantial treatment changes, for cancer surveillance, and when symptoms may not be

disease-related, and if fecal calprotectin is high; however, it is not usually indicated during

less severe relapses.

Statement 107: If fecal calprotectinFC is accessible, it should be collected when in clinical

remission, and endoscopic examination should be undertaken when calprotectin is high.

Vote: 100% strongly agreed; high-quality evidence

Ileo-colonoscopy has proven its efficacy as a diagnostic tool of IBD in children in

many studies. It allows initial and differential diagnosis, and, even though gastrointestinal

malignancies are rare in the pediatric group, could be used for cancer surveillance, especially

after treatment with possible teratogenic drugs (235-237).

High fecal calprotectin levels correlate well with endoscopic disease activity, so it is

advisable to perform an ileo-colonoscopy with biopsy when this marker is increased (238).

Medical Management of UC
ASA and Enemas

Statement 108: For mild-to-moderate UC, oral 5-ASA is suggested as first-line approach to

induceitiate and maintain clinical remission. Rectal and oral 5-ASA combination treatment is

highly effective than using oral 5-ASA alone.

Vote: 100% strongly agreed; high-quality evidence

60
Current evidence supports the use of 5-ASA for induction and maintenance of

remission in mild to moderate UC (127, 239).

Mesalamine appears to induces remission in 35% to 55% of children, as defined by

the PUCAI score (240, 241). In another pediatric RCT, low versus higher dose balsalazide

induced remission in 9%versus 12% of children, respectively (242). Combining oral and

rectal 5-ASA therapy appears to improve clinical outcomes (243, 244).

Statement 109: Rectal monotherapy must be used only in case of mild-to-moderate

ulcerative proctitis. When rectal treatment is chosen, 5-ASA is preferable over

corticosteroids.

Vote: 100% strongly agreed; moderate-quality evidence

Rectal therapy (as suppositories) is indicated for ulcerative proctitis, an infrequent

phenotype in pediatrics (245). In a pediatric ulcerative proctitis trial, mesalamine

suppositories (0.5 g daily) were associated with improved disease activity at 3 and 6 weeks in

children with mild-moderate proctitis (245).

Oral Corticosteroids

Statement 110: Oral corticosteroids must be administered as a second-line treatment in mild-

moderate UC with no response to 5-ASA treatments, and as a first-line treatment for patient

presenting at the higher end of the moderate disease severity spectrum.

Vote: 100% strongly agreed; moderate-quality evidence

Several studies that evaluated the use of oral steroids for treating children with active

UC reported short-term remission rates of 50% to 64% (246, 247).

In a prospective study, that evaluated clinical and histological outcome in patients

treated for 8 weeks with steroids or 5-ASA, the authors reported that 87% had clinical

61
remission, 40% of patients achieved endoscopic remission, and 15% of patients obtained

histological remission (248).

Statement 111: In children with mild disease not responding to 5-ASA, second-generation

oral steroids such as budesonide-MMX and beclomethasone dipropionate (BDP) may be tried

before oral prednisolone. The use of budesonide-MMX is preferable for left-sided colitis.

Vote: 100% strongly agreed; moderate-quality evidence

Second-generation steroids may be considered before systemic steroids in selected

patients (112).

Studies in adults demonstrate the effectiveness of BDP compared with both

prednisolone and mesalamine (185, 249), but there is limited data regarding its efficacy in

pediatric patients.

An RCT that evaluated children with mild-to-moderate UC showed that oral BDP, 5

mg/day for 4 weeks, was more effective than 5- ASA in achieving both clinical and

endoscopic remission (250).

As for budesonide-MMX, it showed minimal clinical effectiveness in a case series

study of children with pancolitis (251).

Statement 112: Corticosteroids should not be used to maintain remission; instead,

corticosteroid-sparing medications should be used in steroid dependent children or frequently

relapsing disease.

Vote: 100% strongly agreed; moderate-quality evidence

Higher steroid dependency rates were reported in pediatric population when compared

to adults (45% vs 8%, respectively) (252), so it is important to develop an individualized

pediatric management. Strategies to avoid steroid dependency include optimization of 5-

ASA, adjuvant therapy with enemas, and a change of therapy to thiopurines or biologic

agents.

62
Immunosuppressants

Statement 113: Thiopurines are recommended as maintenance therapy corticosteroid-sparing

agents for corticosteroid-dependent or frequently relapsing children with UC (≥ 2 relapses

yearly) while receiving optimum 5-ASA therapy. Thiopurines should be considered after

discharge following an episode of severe acute colitis.

Vote: 100% strongly agreed; low-quality evidence

Few studied have evaluated the efficacy of thiopurines for both induction and

maintenance o

of remission in adult UC patients. A few prospective pediatric trials reported steroid-free

remission rates of 49% at 1 year (253) and 72% at 2 years (254) in thiopurine-treated

children, with no difference in either clinical or endoscopic end-points between early or late

initiation of treatment. A few retrospective studies (255, 256) in children supported the

benefit of thiopurines in maintaining remission and steroid sparing, with a median time to

achieve steady state of thiopurine levels of 55 days (257).

Statement 114: Induction of remission with thiopurines is not recommended in children with

UC.

Vote: 100% strongly agreed; low-quality evidence

Limited data does not support the use of thiopurines as an induction of remission

agents in children due to low effectiveness (134, 135, 258).

Statement 115: Patients with an inadequate response to steady thiopurine dose, leucopenia or

increased transaminases, should have their thiopurine metabolites measured.

Vote: 50% strongly agreed; low-quality evidence

63
Several studies supported the idea of testing for TMPT in order to prevent

pancytopenia, or more specifically to avoid leucopenia, and to be able to use aggressive

dosing (259-261).

Biologics

Statement 116: We recommend using Infliximab to induce and maintain remission in

chronically active or corticosteroid-dependent UC pediatric patients resistant to 5-ASA and

thiopurines.

Vote: 100% strongly agreed; low-quality evidence

A study evaluated the response to a standard induction protocol of infliximab of

children with moderate-severe UC (262). The response rate to infliximab was 75% (45/ of 60

patients). Both clinical remission (PUCAI < 10 points) and complete mucosal healing (Mayo

endoscopic subscore 0) were achieved in 33% at week 8. Dose-escalation to 10 mg/kg was

required in almost half of the patients (44%) in the maintenance phase.

Different studies in children have shown a pooled long-term success rate of IFX in

UC of 64% (263), and a corticosteroid-free remission of 38% and 21% at 12 and 24 months,

respectively (253).

Statement 116: Based on serum drug levels and anti-drug antibodyADA concentration,

adalimumab or golimumab could be used as alternative in patients who loset response to

infliximab (secondary non response) or are intolerant to infliximab. In patients who lack

response to biological therapy (primary non-responsePNR), vedolizumab is recommended as

a second-line biological therapy after anti-TNF alpha failure.

Vote: 100% strongly agreed; moderate-quality evidence

64
A meta-analysis that evaluated the TNF inhibitors for moderate-to-severe adult UC

suggested that while infliximab is more effective than adalimumab in the induction of

remission, response and mucosal healing, both are comparable in efficacy (264).

In a retrospective study the authors demonstrated that 60% of UC children who

discontinued infliximab were commenced on adalimumab, with 83% of these remaining on

adalimumab at last follow-up (265). In another retrospective study, 55% of UC children

switched to adalimumab after infliximab failure, achieved and maintained clinical remission

at a median of 25 months (266).

Golimumab has been studied in an open-label pharmacokinetic study of 35 children

with moderate-severe UC (267). Among week 6 Mayo clinical responders (60%) who

continued to receive 4 weekly golimumab maintenance, 57% were in PUCAI remission at

week 14.

A recent study that evaluated the effectiveness of anti-TNF agents compared to

vedolizumab as second-line biologics in IBD patients concluded that the effectiveness and

safety of second-line anti-TNF and vedolizumab at 12 months appeared largely similar (268).

Nonetheless, drug and antibody levels should dictate the course of action in patients

with secondary loss of response. Ongoing symptoms despite adequate drug levels, mandates

switching therapy, while high antibodies titer predicts failure of dose intensification (269).

Statement 117

General considerations for using Biologics


Statement 118

Statement 119: Patients on anti- TNF therapy (infliximab and adalimumab) with a high anti-

drug antibodyADA level should be switched to another biologic therapy within the same

treatment class when available. In the absence of alternatives from the same class, switching

to a different biologic class is possible.

65
Vote: 100% strongly agreed; high-quality evidence

Vedolizumab was gained recognition as a treatment option for moderate to severe UC

and CD in patients who have failed at least 1 other agent. The GEMINI 3 study of

vedolizumab in patients with CD who had previously failed at least 1 other anti-TNF α agent

found that at week 10 of treatment, a higher proportion of patients given vedolizumab were in

remission (26.6%) than patients given placebo (270).

Afif et al., reported that among patients who tested positive for antibodies to

infliximab, 92% (11/12) had a complete or partial response after changing to another anti-

TNF α agent (271).

Several studies support the idea of switching to a different biologic class if another

anti- TNF agent is unavailable, and the patients have high anti-drug antibodyADA levels

(272, 273).

Statement 120: During biological therapy, patients should be monitored for TB contact

history, suggestive manifestations, and indications of active TB. Chest x-ray and IGRA may

be performed yearly.

Vote: 100% strongly agreed; high-quality evidence

Pulmonary tTuberculosis is considered an absolute contraindication for biologic

therapy (96), as suchso it must be excluded before the treatment’s initiation using standard

diagnosis protocol, which includes chest radiography and a purified protein derivative (PPD)

skin test and/or an interferon-gamma release assay (IGRA).

Statement 121: We suggest AGAINST stopping biological if the child is in clinical

remission. We recommend children who undergo relapse after discontinuing treatment to

resume their biological therapy treatment.

Vote: 100% strongly agreed; high-quality evidence

66
Pediatric patients in clinical, biochemical, and endoscopic remission are more likely

to remain well when anti-TNF or immunomodulators immunosuppressants are stopped.

Reintroduction of the same treatment is usually, but not always, successful and close clinical

monitoring is required upon any treatment withdrawal (274).

The decision on treatment withdrawal is also based on patient preference. Patients

with subclinical disease activity are at much higher risk of relapse when any treatment is

reduced or withdrawn (275).

Before withdrawal of any maintenance IBD therapy, re-evaluation of disease activity

using a combination of clinical, biochemical, endoscopic/histological, and/or radiological

techniques should be performed to assess risks and benefits of stopping (147).

Statement 122

Statement 123: IBD patients should be tested for hepatitis B virus, hepatitis C virus, and

varicella-zoster virus (without any history of varicella immunization, chicken pox or shingles

chickenpox, shingles, or varicella immunization) before starting immunosuppressants or

biological therapy.

Vote: 100% strongly agreed; high-quality evidence

To minimize the risk of viral reactivation, immunosuppressive therapy should proceed

only after screening of hepatitis B, hepatitis C, and varicella-zoster viruses, in order to avoid

life threateninglife-threatening situations (276, 277). HBV vaccination is recommended in

patients who are negative for HBsAg, HBsAb, and HBcAb.

Medical management of Crohn’s disease in pediatrics (CD)


Aminosalicylates

Statement 124: We recommend AGAINST using 5-ASA to induce clinical remission in

children with moderate CD.

67
Vote: 100% strongly agreed; high-quality evidence

Several studies provided data on the use of 5-ASA in mild to moderate active CD

(278-280). These analyses included studies using various formulations and doses of non-

sulfasalazine 5-ASAs (i.e., mesalamine and olsalazine), and generally reported no benefit

with these agents over placebo for induction of remission.

The data did not assess mild and moderate disease separately, and no RCTs in

pediatric patients were found, so it is reasonable not to use 5-ASA agents to induce clinical

remission in children with moderate CD.

Statement 125: We suggest AGAINST using sulfasalazine to encourage clinical remission in

children with moderate CD confined to the colon.

Vote: 100% strongly agreed; low-quality evidence

The data is scarce regarding the efficacy of sulfasalazine in CD. Some studies have

indicated that sulfasalazine has a marginal benefit over placebo from a clinical remission

point of view (278-280).

No RCTs were found in pediatric patients, and while mild disease was analyzed

separately, the subgroup of patients was very small.

Statement 126: We recommend AGAINST using 5-ASA or sulfasalazine to maintain

remission in children with CD.

Vote: 100% strongly agreed; moderate-quality evidence

Two meta-analyses evaluated sulfasalazine and mesalamine for maintenance of

remission of CD (278, 281). Sulfasalazine was not effective in preventing relapse of CD, but

there was a non-significant trend toward improvement over placebo with mesalamine.

A pediatric RCT involving 132 patients reported no statistically significant difference

in relapse rates at one year with mesalamine compared to placebo (282).

68
Corticosteroids

Statement 127: We recommend use of conventional corticosteroids to trigger clinical

remission in IBD children with moderate-to-severe CD.

Vote: 100% strongly agreed; moderate-quality evidence

Corticosteroids are effective for the induction of clinical remission in CD and UC in

children, but approximately half of the patients will become dependent on corticosteroids or

require surgery (246).

Statement 128: We suggest use of conventional corticosteroids to encourage clinical

remission in children with mild-to-moderate active CD who failed 5-ASA, sulfasalazine, oral

budesonide, or did not respond to exclusive enteral nutrition.

Vote: 100% strongly agreed; moderate-quality evidence

Corticosteroids continue to be the basic treatment to control acute disease that has not

responded to first line therapy (i.e., 5-ASA) (283). Conventional corticosteroid therapy is

commonly used for the short-term treatment of moderate to severe symptoms, and is typically

used for achieving a quick relief of symptoms. It is important to minimize the use of

corticosteroids as long-term use results in adverse effects.

As for the failure of exclusive enteral nutrition, the data is scarce, but it seems that

corticosteroids help with the clinical remission in patients with CD (284).

Data from several small studies comparing treatment with prednisone to budesonide

illustrated no superiority of budesonide over traditional corticosteroids (285-287). In fact,

prednisone may be more effective for inducing remission in patients who failed budesonide

therapy.

69
Statement 129: We suggest AGAINST use of corticosteroids to maintain clinical remission

in children with CD, regardless of severity.

Vote: 100% strongly agreed; moderate-quality evidence

A meta-analysis found no significant reduction in the odds of relapse with ongoing

corticosteroid therapy compared to placebo (288). Due to the lack of demonstrated efficacy in

preventing relapse, and concerns around the adverse events associated with long-term use,

particularly in children, we agreed that corticosteroids should not be used to maintain clinical

remission in children with CD, regardless of severity.

Exclusive Enteral Nutrition

Statement 130: We suggest inducing clinical remission in pediatric patients with CD using

exclusive enteral nutrition.

Statement 131: Partial enteral nutrition is not allowed to trigger clinical remission in

pediatric patients with CD. It should only be as an adjunctive to medications that maintain

clinical remission.

Vote: 100% strongly agreed; moderate-quality evidence

In one RCT that evaluated children with active CD, partial enteral nutrition was less

effective than total enteral nutrition for the induction of clinical remission (289). Similarly, in

a recent multicenter North American study, patients allowed 20% of total calories as regular

food actually consumed 50% of calories as such, and were less likely to achieve either

clinical remission or reduction in fecal calprotectin compared with those receiving exclusive

enteral nutrition (290).

132

Immunosuppressants

70
Statement 133: We recommend AGAINST using thiopurine alone to induce clinical

remission in children with CD of any severity. In female patients with CD, we suggest

thiopurine therapy for maintaining remission.

Vote: 100% strongly agreed; high-quality evidence

Two meta-analyses reported no significant improvement in symptomatic remission rates

with thiopurine therapy (azathioprine or 6-mercaptopurine) when compared to placebo (291,

292).

A meta-analysis found that azathioprine was significantly superior to placebo in

maintaining remission over 6–18 months (134). One additional small RCT withdrawal trial,

published after the meta-analyses, also reported a reduction in risk of relapse with ongoing

thiopurine therapy, which was significant at 1 year (293).

A small RCT found that, in children receiving corticosteroid therapy, the addition of

6-mercaptopurine was associated with a lower rate of relapse compared to adjunctive placebo

at 18 months (294).

Statement 134: We suggest thiopurine methyltransferase (TPMT) testing by genotyping or

enzymatic activity in pediatric CD patients if available before starting thiopurine treatment

for dosing determination.

Vote: 100% strongly agreed; moderate-quality evidence

Three RCTs assessed the benefits of thiopurine methyltransferase (TPMT) testing,

using genotyping (295, 296) or enzymatic activity (297), compared to no testing before

initiating thiopurine therapy to individualize dosing. Among the 1145 patients included, only

2 (0.17%) patients were homozygous, and 150 (13.1%) were heterozygous for variant alleles

in the TPMT gene.

While most patients with leukopenia in the largest study did not have reduced

enzymatic activity, individuals with TPMT mutations and low or intermediate enzymatic

71
activity had a significant reduction in the risk of hematologic adverse events with TPMT

testing to guide dosing (295).

Statement 135: Parenteral methotrexate is suggested for use in children with CD as

maintenance treatment. We suggest assessment for mucosal healing in children with CD in

remission using thiopurines or methotrexate within the first year to judge the need for

treatment modification.

Vote: 100% strongly agreed; high-quality evidence

Several studies confirmed the efficacy of parenteral methotrexate for maintenance

therapy in adults with CD in clinical remission (296, 298). In the SR of pediatric

observational studies, long-term remission rates were 37% to 62% at 6 months, and 25% to

53% at 12 months, primarily with parenteral methotrexate (299).

In pediatric patients, complete mucosal healing was associated with significantly

higher rates of long-term remission for up to 3 years compared to ongoing active endoscopic

disease (300). Rates of mucosal healing with anti-TNF therapy (27%‒31%) were higher than

those seen with immunosuppressants (16.5%) or placebo (0‒13%) (301-303).

Anti–Tumor Necrosis Factor Biologic Therapy

Statement 136: We propose anti-TNF treatment (including infliximab and adalimumab) in

children with inflammatory moderate-to-severe CD who have failed induction of clinical

remission with corticosteroids for both induction and maintenance of remission.

Vote: 100% strongly agreed; high-quality evidence

For induction therapy, a meta-analysis found that anti-TNF therapy alone or with

concomitant therapies was significantly more effective than placebo for the outcome of

failure to achieve symptomatic remission (278). Results were significant for infliximab and

adalimumab, but not certolizumab pegol. For maintenance therapy, another meta-analysis

72
showed that anti-TNF therapy significantly reduced the risk of relapse in patients with CD in

clinical remission compared to placebo. Results were significant for infliximab and

certolizumab pegol, but not adalimumab.

In another meta-analysis, all 3 anti-TNFs were significantly more effective than

placebo for both induction and maintenance of remission (304).

Statement 137: We recommend anti-TNF treatment for induction and maintenance of

remission in children with moderate-to-severe inflammatory CD who have failed to respond

to thiopurine or methotrexate.

Statement 138: We suggest anti-TNF therapy as first-line treatment to induce and maintain

clinical remission in children with severe inflammatory CD at higher risk for progressive and

complicated disease.

Vote: 100% strongly agreed; high-quality evidence

Additional support for the early use of anti-TNF therapy comes from two prospective

trials using combined anti-TNF and immunosuppressive therapy in newly diagnosed,

treatment naïve patients (158, 305). In these studies, the “top-down” treatment was associated

with significantly higher rates of symptomatic remission at earlier time points compared to

not using early anti-TNF therapy.

Statement 139: We recommend combining Infliximab with immunosuppressants for the first

4 to 6 months of therapy.

Vote: 100% strongly agreed; high-quality evidence

We agree with the recent European guidelines for treatment of pediatric UC, which

recommend induction therapy with IFX in combination with an immunomodulator, and

discontinuation of the immunomodulator after 6 months, especially in boys (234).

73
Statement 140: We suggest AGAINST combining adalimumab with immunosuppressants in

patients who are bio-naive.

Vote: 100% strongly agreed; high-quality evidence

The DIAMOND RCT evaluated adult patients who were immunosuppressant- and

biologic-naïve, and found no difference in 26-week clinical remission rates between the

combination of adalimumab plus azathioprine (68.1%) and adalimumab monotherapy

(71.8%) (306).

Similarly, post-hoc analyses of cohort data from RCTs in adults did not show a

significant benefit with combination adalimumab and immunosuppressant therapy (thiopurine

or methotrexate) over adalimumab alone for induction or maintenance of remission (307).

Statement 141: We suggest AGAINST using thiopurines in combination with anti-TNF

therapy in male children with CD. We suggest using parenteral methotrexate instead.

Vote: 100% strongly agreed; high-quality evidence

The COMMIT study compared the efficacy of combination therapy with infliximab

plus methotrexate to infliximab alone, and found no difference in rates of symptomatic

remission between the 2 treatment groups (308).

In a RCT which evaluated pediatric patients on either azathioprine or methotrexate in

the combination treatment group (309). Overall combination therapy with infliximab plus an

immunosuppressant was not associated with a benefit over infliximab alone in preventing

loss of response over the 1-year follow-up.

Statement 142: We suggest treatment intensification based on therapeutic drug monitoring

(TDM) in children with CD with poor clinical response to anti-TNF induction treatment or

loss of response to maintenance therapy.

Vote: 100% strongly agreed; high-quality evidence

74
Several studies have demonstrated that antibodies to anti-TNFs were associated with

greater likelihood of loss of response, and higher serum anti-TNF levels were associated with

a greater probability of clinical remission and mucosal healing (310-312).

In a retrospective case series of pediatric patients with IBD, those with very low

infliximab drug levels had high rates of infliximab antibodies, non-response, or loss of

response (313).

Statement 143: Ustekinumab is recommended for children with moderate-to-severe CD who

have not achieved or maintained clinical remission with anti-TNF therapy.

Vote: 100% strongly agreed; high-quality evidence

Two RCTs assessed the use of ustekinumab as maintenance therapy in patients who

previously failed anti-TNF therapy. In the CERTIFI trial, ustekinumab resulted in

significantly increased rates of clinical remission at 22 weeks compared with placebo (41.7%

vs 27.4%; P = .03) (314).

In the combined population in the UNITI-IM trial, significantly more patients were in

remission with maintenance ustekinumab after 1 year of treatment compared to placebo

(49%–53% vs 36%) (41).

Conclusions:

75
References:

1. Gajendran M, Loganathan P, Jimenez G, Catinella AP, Ng N, Umapathy C, et al. A


comprehensive review and update on ulcerative colitis(). Dis Mon. 2019;65(12):100851.
2. Köhne G, Stallmach A, Zeitz M. [Recurrent Crohn disease--definition and diagnosis].
Chirurg. 1995;66(8):745-50.
3. Nikolaus S, Schreiber S. Diagnostics of Inflammatory Bowel Disease.
Gastroenterology. 2007;133(5):1670-89.
4. Maaser C, Sturm A, Vavricka SR, Kucharzik T, Fiorino G, Annese V, et al. ECCO-ESGAR
Guideline for Diagnostic Assessment in IBD Part 1: Initial diagnosis, monitoring of known
IBD, detection of complications. Journal of Crohn's and Colitis. 2018;13(2):144-64K.
5. Gecse KB, Vermeire S. Differential diagnosis of inflammatory bowel disease:
imitations and complications. Lancet Gastroenterol Hepatol. 2018;3(9):644-53.
6. Bhaijee F, Arnold C, Lam-Himlin D, Montgomery EA, Voltaggio L. Infectious mimics of
inflammatory bowel disease. Diagnostic Histopathology. 2015;21(7):267-75.
7. Danese S, Fiorino G, Mary J-Y, Lakatos PL, D’Haens G, Moja L, et al. Development of
Red Flags Index for Early Referral of Adults with Symptoms and Signs Suggestive of Crohn’s
Disease: An IOIBD Initiative. Journal of Crohn's and Colitis. 2015;9(8):601-6.
8. Mosli M, Bamarhul M, Alharbi A, Shafei S, Alharbi A, Bamahfouth K, et al. Screening
irritable bowel syndrome patients for symptoms predictive of crohn's disease using the red
flag score. Saudi J Gastroenterol. 2017;23(4):229-32.
9. Bossuyt X. Serologic markers in inflammatory bowel disease. Clinical chemistry.
2006;52(2):171-81.
10. Reese GE, Constantinides VA, Simillis C, Darzi AW, Orchard TR, Fazio VW, et al.
Diagnostic precision of anti-Saccharomyces cerevisiae antibodies and perinuclear
antineutrophil cytoplasmic antibodies in inflammatory bowel disease. Am J Gastroenterol.
2006;101(10):2410-22.

76
11. Chen P, Zhou G, Lin J, Li L, Zeng Z, Chen M, et al. Serum Biomarkers for Inflammatory
Bowel Disease. Front Med (Lausanne). 2020;7:123.
12. Mirkov MU, Verstockt B, Cleynen I. Genetics of inflammatory bowel disease: beyond
NOD2. The lancet Gastroenterology & hepatology. 2017;2(3):224-34.
13. Gisbert JP, Bermejo F, Pérez-Calle J-L, Taxonera C, Vera I, McNicholl AG, et al. Fecal
calprotectin and lactoferrin for the prediction of inflammatory bowel disease relapse.
Inflammatory bowel diseases. 2009;15(8):1190-8.
14. Von Roon AC, Karamountzos L, Purkayastha S, Reese GE, Darzi AW, Teare JP, et al.
Diagnostic precision of fecal calprotectin for inflammatory bowel disease and colorectal
malignancy. Official journal of the American College of Gastroenterology| ACG.
2007;102(4):803-13.
15. Schoepfer AM, Beglinger C, Straumann A, Trummler M, Renzulli P, Seibold F.
Ulcerative colitis: correlation of the Rachmilewitz endoscopic activity index with fecal
calprotectin, clinical activity, C-reactive protein, and blood leukocytes. Inflammatory bowel
diseases. 2009;15(12):1851-8.
16. Vinding KK, Elsberg H, Thorkilgaard T, Belard E, Pedersen N, Elkjaer M, et al. Fecal
calprotectin measured by patients at home using smartphones—a new clinical tool in
monitoring patients with inflammatory bowel disease. Inflammatory bowel diseases.
2016;22(2):336-44.
17. Langner C, Magro F, Driessen A, Ensari A, Mantzaris GJ, Villanacci V, et al. The
histopathological approach to inflammatory bowel disease: a practice guide. Virchows
Archiv. 2014;464(5):511-27.
18. Lin WC, Chang CW, Chen MJ, Hsu TC, Wang HY. Effectiveness of sigmoidoscopy for
assessing ulcerative colitis disease activity and therapeutic response. Medicine (Baltimore).
2019;98(21):e15748.
19. Spiceland CM, Lodhia N. Endoscopy in inflammatory bowel disease: Role in
diagnosis, management, and treatment. World J Gastroenterol. 2018;24(35):4014-20.
20. Passos MAT, Chaves FC, Chaves-Junior N. THE IMPORTANCE OF COLONOSCOPY IN
INFLAMMATORY BOWEL DISEASES. Arq Bras Cir Dig. 2018;31(2):e1374-e.
21. Magro F, Langner C, Driessen A, Ensari A, Geboes K, Mantzaris GJ, et al. European
consensus on the histopathology of inflammatory bowel disease☆. Journal of Crohn's and
Colitis. 2013;7(10):827-51.
22. Bitton A, Peppercorn MA, Antonioli DA, Niles JL, Shah S, Bousvaros A, et al. Clinical,
biological, and histologic parameters as predictors of relapse in ulcerative colitis.
Gastroenterology. 2001;120(1):13-20.
23. Flynn S, Eisenstein S. Inflammatory Bowel Disease Presentation and Diagnosis. Surg
Clin North Am. 2019;99(6):1051-62.
24. Liu W, Liu J, Xiao W, Luo G. A Diagnostic Accuracy Meta-analysis of CT and MRI for
the Evaluation of Small Bowel Crohn Disease. Academic Radiology. 2017;24(10):1216-25.
25. Gallego JC, Echarri A. Role of magnetic resonance imaging in the management of
perianal Crohn's disease. Insights Imaging. 2018;9(1):47-58.
26. Sahni VA, Ahmad R, Burling D. Which method is best for imaging of perianal fistula?
Abdom Imaging. 2008;33(1):26-30.
27. Raman SP, Horton KM, Fishman EK. Computed tomography of Crohn's disease: The
role of three dimensional technique. World J Radiol. 2013;5(5):193-201.
28. Calabrese E, Maaser C, Zorzi F, Kannengiesser K, Hanauer SB, Bruining DH, et al.
Bowel Ultrasonography in the Management of Crohn's Disease. A Review with

77
Recommendations of an International Panel of Experts. Inflamm Bowel Dis.
2016;22(5):1168-83.
29. Magro F, Gionchetti P, Eliakim R, Ardizzone S, Armuzzi A, Barreiro-de Acosta M, et al.
Third European Evidence-based Consensus on Diagnosis and Management of Ulcerative
Colitis. Part 1: Definitions, Diagnosis, Extra-intestinal Manifestations, Pregnancy, Cancer
Surveillance, Surgery, and Ileo-anal Pouch Disorders. Journal of Crohn's and Colitis.
2017;11(6):649-70.
30. Fine KD, Schiller LR. AGA technical review on the evaluation and management of
chronic diarrhea. Gastroenterology. 1999;116(6):1464-86.
31. D'Aoust J, Battat R, Bessissow T. Management of inflammatory bowel disease with
Clostridium difficile infection. World J Gastroenterol. 2017;23(27):4986-5003.
32. Mylonaki M, Langmead L, Pantes A, Johnson F, Rampton DS. Enteric infection in
relapse of inflammatory bowel disease: importance of microbiological examination of stool.
Eur J Gastroenterol Hepatol. 2004;16(8):775-8.
33. Masclee GM, Penders J, Jonkers DM, Wolffs PF, Pierik MJ. Is clostridium difficile
associated with relapse of inflammatory bowel disease? results from a retrospective and
prospective cohort study in the Netherlands. Inflamm Bowel Dis. 2013;19(10):2125-31.
34. Gryboski JD. Clostridium difficile in inflammatory bowel disease relapse. J Pediatr
Gastroenterol Nutr. 1991;13(1):39-41.
35. Berg AM, Kelly CP, Farraye FA. Clostridium difficile infection in the inflammatory
bowel disease patient. Inflamm Bowel Dis. 2013;19(1):194-204.
36. Heidari H, Sedigh Ebrahim-Saraie H, Amanati A, Motamedifar M, Hadi N, Bazargani A.
Toxin profiles and antimicrobial resistance patterns among toxigenic clinical isolates of
Clostridioides (Clostridium) difficile. Iran J Basic Med Sci. 2019;22(7):813-9.
37. Leighton JA, Shen B, Baron TH, Adler DG, Davila R, Egan JV, et al. ASGE guideline:
endoscopy in the diagnosis and treatment of inflammatory bowel disease. Gastrointestinal
endoscopy. 2006;63(4):558-65.
38. Kedia S, Das P, Madhusudhan KS, Dattagupta S, Sharma R, Sahni P, et al.
Differentiating Crohn's disease from intestinal tuberculosis. World J Gastroenterol.
2019;25(4):418-32.
39. Liu Y-Y, Chen M-K, Cao Z, Liu S-Z, Ding B-J. Differential diagnosis of intestinal
tuberculosis from Crohn's disease and primary intestinal lymphoma in China. Saudi J
Gastroenterol. 2014;20(4):241-7.
40. Du J, Ma Y-Y, Xiang H, Li Y-M. Confluent granulomas and ulcers lined by epithelioid
histiocytes: new ideal method for differentiation of ITB and CD? A meta analysis. PLoS One.
2014;9(10):e103303-e.
41. Feagan BG, Sandborn WJ, Gasink C, Jacobstein D, Lang Y, Friedman JR, et al.
Ustekinumab as Induction and Maintenance Therapy for Crohn's Disease. N Engl J Med.
2016;375(20):1946-60.
42. Bae J, Park sh, Ye B, Kim S, Cho Y, Youn E, et al. Development and Validation of a
Novel Prediction Model for Differential Diagnosis Between Crohn's Disease and Intestinal
Tuberculosis. Inflammatory bowel diseases. 2017;23.
43. Jin XJ, Kim JM, Kim HK, Kim L, Choi SJ, Park IS, et al. Histopathology and TB-PCR kit
analysis in differentiating the diagnosis of intestinal tuberculosis and Crohn's disease. World
J Gastroenterol. 2010;16(20):2496-503.

78
44. Annunziata ML, Caviglia R, Papparella LG, Cicala M. Upper gastrointestinal
involvement of Crohn’s disease: a prospective study on the role of upper endoscopy in the
diagnostic work-up. Digestive diseases and sciences. 2012;57(6):1618-23.
45. Rutgeerts P, Onette E, Vantrappen G, Geboes K, Broeckaert L, Talloen L. Crohn's
disease of the stomach and duodenum: a clinical study with emphasis on the value of
endoscopy and endoscopic biopsies. Endoscopy. 1980;12(06):288-94.
46. Kopylov U, Yung DE, Engel T, Vijayan S, Har-Noy O, Katz L, et al. Diagnostic yield of
capsule endoscopy versus magnetic resonance enterography and small bowel contrast
ultrasound in the evaluation of small bowel Crohn’s disease: systematic review and meta-
analysis. Digestive and Liver Disease. 2017;49(8):854-63.
47. Triester SL, Leighton JA, Leontiadis GI, Gurudu SR, Fleischer DE, Hara AK, et al. A
meta-analysis of the yield of capsule endoscopy compared to other diagnostic modalities in
patients with non-stricturing small bowel Crohn's disease. Official journal of the American
College of Gastroenterology| ACG. 2006;101(5):954-64.
48. Van Weyenberg SJ, Bouman K, Jacobs MA, Halloran BP, Van der Peet DL, Mulder CJ,
et al. Comparison of MR enteroclysis with video capsule endoscopy in the investigation of
small-intestinal disease. Abdominal imaging. 2013;38(1):42-51.
49. Nemeth A, Kopylov U, Koulaouzidis A, Johansson GW, Thorlacius H, Amre D, et al.
Use of patency capsule in patients with established Crohn’s disease. Endoscopy.
2016;48(04):373-9.
50. Mow WS, Lo SK, Targan SR, Dubinsky MC, Treyzon L, Abreu-Martin MT, et al. Initial
experience with wireless capsule enteroscopy in the diagnosis and management of
inflammatory bowel disease. Clinical Gastroenterology and Hepatology. 2004;2(1):31-40.
51. Monteiro S, de Castro FD, Carvalho PB, Rosa B, Moreira MJ, Pinho R, et al. Essential
role of small bowel capsule endoscopy in reclassification of colonic inflammatory bowel
disease type unclassified. World journal of gastrointestinal endoscopy. 2017;9(1):34.
52. Hellers G, Bergstrand O, Ewerth S, Holmström B. Occurrence and outcome after
primary treatment of anal fistulae in Crohn's disease. Gut. 1980;21(6):525-7.
53. Sandborn WJ, Fazio VW, Feagan BG, Hanauer SB. AGA technical review on perianal
Crohn’s disease. Gastroenterology. 2003;125(5):1508-30.
54. Regueiro M. The role of endoscopy in the evaluation of fistulizing Crohn's disease.
Gastrointestinal Endoscopy Clinics. 2002;12(3):621-33.
55. Silverberg MS, Satsangi J, Ahmad T, Arnott ID, Bernstein CN, Brant SR, et al. Toward
an integrated clinical, molecular and serological classification of inflammatory bowel
disease: report of a Working Party of the 2005 Montreal World Congress of
Gastroenterology. Can J Gastroenterol. 2005;19 Suppl A:5a-36a.
56. Spekhorst LM, Visschedijk MC, Alberts R, Festen EA, van der Wouden E-J, Dijkstra G,
et al. Performance of the Montreal classification for inflammatory bowel diseases. World J
Gastroenterol. 2014;20(41):15374-81.
57. Levine A, Griffiths A, Markowitz J, Wilson DC, Turner D, Russell RK, et al. Pediatric
modification of the Montreal classification for inflammatory bowel disease: the Paris
classification. Inflamm Bowel Dis. 2011;17(6):1314-21.
58. Dotson JL, Crandall WV, Zhang P, Forrest CB, Bailey LC, Colletti RB, et al. Feasibility
and validity of the pediatric ulcerative colitis activity index in routine clinical practice. J
Pediatr Gastroenterol Nutr. 2015;60(2):200-4.

79
59. Zittan E, Kabakchiev B, Kelly OB, Milgrom R, Nguyen GC, Croitoru K, et al.
Development of the Harvey-Bradshaw Index-pro (HBI-PRO) Score to Assess Endoscopic
Disease Activity in Crohn's Disease. J Crohns Colitis. 2017;11(5):543-8.
60. Harvey RF, Bradshaw JM. A simple index of Crohn's-disease activity. Lancet.
1980;1(8167):514.
61. Daperno M, D'Haens G, Van Assche G, Baert F, Bulois P, Maunoury V, et al.
Development and validation of a new, simplified endoscopic activity score for Crohn's
disease: the SES-CD. Gastrointest Endosc. 2004;60(4):505-12.
62. Lewis JD, Chuai S, Nessel L, Lichtenstein GR, Aberra FN, Ellenberg JH. Use of the
noninvasive components of the Mayo score to assess clinical response in ulcerative colitis.
Inflamm Bowel Dis. 2008;14(12):1660-6.
63. Turner D, Otley AR, Mack D, Hyams J, de Bruijne J, Uusoue K, et al. Development,
validation, and evaluation of a pediatric ulcerative colitis activity index: a prospective
multicenter study. Gastroenterology. 2007;133(2):423-32.
64. Turner D, Seow CH, Greenberg GR, Griffiths AM, Silverberg MS, Steinhart AH. A
systematic prospective comparison of noninvasive disease activity indices in ulcerative
colitis. Clin Gastroenterol Hepatol. 2009;7(10):1081-8.
65. Turner D, Mack D, Leleiko N, Walters TD, Uusoue K, Leach ST, et al. Severe pediatric
ulcerative colitis: a prospective multicenter study of outcomes and predictors of response.
Gastroenterology. 2010;138(7):2282-91.
66. Hyams JS, Ferry GD, Mandel FS, Gryboski JD, Kibort PM, Kirschner BS, et al.
Development and validation of a pediatric Crohn's disease activity index. J Pediatr
Gastroenterol Nutr. 1991;12(4):439-47.
67. Shaoul R, Sladek M, Turner D, Paeregaard A, Veres G, Wauters GV, et al. Limitations
of fecal calprotectin at diagnosis in untreated pediatric Crohn's disease. Inflamm Bowel Dis.
2012;18(8):1493-7.
68. Otley A, Loonen H, Parekh N, Corey M, Sherman PM, Griffiths AM. Assessing activity
of pediatric Crohn's disease: which index to use? Gastroenterology. 1999;116(3):527-31.
69. Grant A, Lerer T, Griffiths AM, Hyams JS, Otley A. Assessing disease activity using the
pediatric Crohn's disease activity index: Can we use subjective or objective parameters
alone? World J Gastroenterol. 2021;27(30):5100-11.
70. Higgins P, Schwartz M, Mapili J, Krokos I, Leung J, Zimmermann E. Patient defined
dichotomous end points for remission and clinical improvement in ulcerative colitis. Gut.
2005;54(6):782-8.
71. Samaan MA, Mosli MH, Sandborn WJ, Feagan BG, D'Haens GR, Dubcenco E, et al. A
systematic review of the measurement of endoscopic healing in ulcerative colitis clinical
trials: recommendations and implications for future research. Inflammatory bowel diseases.
2014;20(8):1465-71.
72. Neurath MF, Travis SP. Mucosal healing in inflammatory bowel diseases: a
systematic review. Gut. 2012;61(11):1619-35.
73. Bryant RV, Costello SP, Schoeman S, Sathananthan D, Knight E, Lau SY, et al. Limited
uptake of ulcerative colitis “treat‐to‐target” recommendations in real‐world practice.
Journal of gastroenterology and hepatology. 2018;33(3):599-607.
74. Bryant R, Winer S, SPL T, Riddell R. Systematic review: histological remission in
inflammatory bowel disease. Is ‘complete’remission the new treatment paradigm? An IOIBD
initiative. Journal of Crohn's and Colitis. 2014;8(12):1582-97.

80
75. Ardizzone S, Cassinotti A, Duca P, Mazzali C, Penati C, Manes G, et al. Mucosal
healing predicts late outcomes after the first course of corticosteroids for newly diagnosed
ulcerative colitis. Clinical Gastroenterology and Hepatology. 2011;9(6):483-9. e3.
76. Meucci G, Fasoli R, Saibeni S, Valpiani D, Gullotta R, Colombo E, et al. Prognostic
significance of endoscopic remission in patients with active ulcerative colitis treated with
oral and topical mesalazine: a prospective, multicenter study. Inflammatory bowel diseases.
2012;18(6):1006-10.
77. Peyrin-Biroulet L, Sandborn W, Sands B, Reinisch W, Bemelman W, Bryant R, et al.
Selecting therapeutic targets in inflammatory bowel disease (STRIDE): determining
therapeutic goals for treat-to-target. Official journal of the American College of
Gastroenterology| ACG. 2015;110(9):1324-38.
78. Turner D, Ricciuto A, Lewis A, D'Amico F, Dhaliwal J, Griffiths AM, et al. STRIDE-II: An
Update on the Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE)
Initiative of the International Organization for the Study of IBD (IOIBD): Determining
Therapeutic Goals for Treat-to-Target strategies in IBD. Gastroenterology.
2021;160(5):1570-83.
79. Hyams J, Markowitz J, Otley A, Rosh J, Mack D, Bousvaros A, et al. Evaluation of the
pediatric crohn disease activity index: a prospective multicenter experience. Journal of
pediatric gastroenterology and nutrition. 2005;41(4):416-21.
80. Kundhal P, Critch J, Zachos M, Otley A, Stephens D, Griffiths A. Pediatric Crohn
Disease Activity Index: responsive to short-term change. Journal of pediatric
gastroenterology and nutrition. 2003;36(1):83-9.
81. Fabiszewska S, Derda E, Szymanska E, Osiecki M, Kierkus J. Safety and Effectiveness
of Vedolizumab for the Treatment of Pediatric Patients with Very Early Onset Inflammatory
Bowel Diseases. J Clin Med. 2021;10(13).
82. Aloi M, Di Nardo G, Conte F, Mazzeo L, Cavallari N, Nuti F, et al. Methotrexate in
paediatric ulcerative colitis: a retrospective survey at a single tertiary referral centre.
Alimentary Pharmacology & Therapeutics. 2010;32(8):1017-22.
83. Tajiri H, Arai K, Kagimoto S, Kunisaki R, Hida N, Sato N, et al. Infliximab for pediatric
patients with ulcerative colitis: a phase 3, open-label, uncontrolled, multicenter trial in
Japan. BMC Pediatr. 2019;19(1):351.
84. Haisma S-M, Verkade HJ, Scheenstra R, van der Doef HPJ, Bodewes FAJA, van
Rheenen PF. Time-to-reach Target Calprotectin Level in Newly Diagnosed Patients With
Inflammatory Bowel Disease. Journal of Pediatric Gastroenterology and Nutrition.
2019;69(4):466-73.
85. Heida A, Park KT, van Rheenen PF. Clinical Utility of Fecal Calprotectin Monitoring in
Asymptomatic Patients with Inflammatory Bowel Disease: A Systematic Review and Practical
Guide. Inflamm Bowel Dis. 2017;23(6):894-902.
86. Boschetti G, Garnero P, Moussata D, Cuerq C, Préaudat C, Duclaux-Loras R, et al.
Accuracies of serum and fecal S100 proteins (calprotectin and calgranulin C) to predict the
response to TNF antagonists in patients with Crohn's disease. Inflamm Bowel Dis.
2015;21(2):331-6.
87. Guidi L, Marzo M, Andrisani G, Felice C, Pugliese D, Mocci G, et al. Faecal calprotectin
assay after induction with anti-Tumour Necrosis Factor α agents in inflammatory bowel
disease: Prediction of clinical response and mucosal healing at one year. Dig Liver Dis.
2014;46(11):974-9.

81
88. Ripollés T, Paredes JM, Martínez-Pérez MJ, Rimola J, Jauregui-Amezaga A, Bouzas R,
et al. Ultrasonographic changes at 12 weeks of anti-TNF drugs predict 1-year sonographic
response and clinical outcome in Crohn's disease: a multicenter study. Inflammatory bowel
diseases. 2016;22(10):2465-73.
89. Kucharzik T, Wittig BM, Helwig U, Börner N, Rössler A, Rath S, et al. Use of intestinal
ultrasound to monitor Crohn’s disease activity. Clinical Gastroenterology and Hepatology.
2017;15(4):535-42. e2.
90. Bruining DH, Loftus Jr EV, Ehman EC, Siddiki HA, Nguyen DL, Fidler JL, et al.
Computed tomography enterography detects intestinal wall changes and effects of
treatment in patients with Crohn's disease. Clinical Gastroenterology and Hepatology.
2011;9(8):679-83. e1.
91. Stoppino LP, Della Valle N, Rizzi S, Cleopazzo E, Centola A, Iamele D, et al. Magnetic
resonance enterography changes after antibody to tumor necrosis factor (anti-TNF) alpha
therapy in Crohn’s disease: correlation with SES-CD and clinical-biological markers. BMC
medical imaging. 2016;16(1):1-9.
92. Shah SC, Colombel J-F, Sands BE, Narula N. Mucosal healing is associated with
improved long-term outcomes of patients with ulcerative colitis: a systematic review and
meta-analysis. Clinical gastroenterology and hepatology. 2016;14(9):1245-55. e8.
93. Orlando A, Mocciaro F, Renna S, Scimeca D, Rispo A, Scribano ML, et al. Early post-
operative endoscopic recurrence in Crohn's disease patients: Data from an Italian Group for
the study of inflammatory bowel disease (IG-IBD) study on a large prospective multicenter
cohort. Journal of Crohn's and Colitis. 2014;8(10):1217-21.
94. Lobatón T, Bessissow T, Ruiz-Cerulla A, De Hertogh G, Bisschops R, Guardiola J, et al.
Prognostic value of histological activity in patients with ulcerative colitis in deep remission: A
prospective multicenter study. United European gastroenterology journal. 2018;6(5):765-72.
95. Arkteg CB, Wergeland Sørbye S, Buhl Riis L, Dalen SM, Florholmen J, Goll R. Real-life
evaluation of histologic scores for Ulcerative Colitis in remission. PLoS One.
2021;16(3):e0248224.
96. Beaugerie L, Rahier JF, Kirchgesner J. Predicting, Preventing, and Managing
Treatment-Related Complications in Patients With Inflammatory Bowel Diseases. Clin
Gastroenterol Hepatol. 2020;18(6):1324-35.e2.
97. Piotrowska M, Krajewska J, Fichna J, Majchrzak K. [Strategies in Crohn's disease
treatment - "step-up" vs. "top-down"]. Postepy Biochem. 2020;65(4):313-7.
98. Rogler G. Top-down or step-up treatment in Crohn's disease? Dig Dis. 2013;31(1):83-
90.
99. Tsui JJ, Huynh HQ. Is top-down therapy a more effective alternative to conventional
step-up therapy for Crohn's disease? Ann Gastroenterol. 2018;31(4):413-24.
100. Holvoet T, Lobaton T, Hindryckx P. Optimal Management of Acute Severe Ulcerative
Colitis (ASUC): Challenges and Solutions. Clin Exp Gastroenterol. 2021;14:71-81.
101. Verdon C, Bessissow T, Lakatos PL. Management of Acute Severe Colitis in the Era of
Biologicals and Small Molecules. J Clin Med. 2019;8(12):2169.
102. Seah D, De Cruz P. Review article: the practical management of acute severe
ulcerative colitis. Alimentary Pharmacology & Therapeutics. 2016;43(4):482-513.
103. Torres J, Bonovas S, Doherty G, Kucharzik T, Gisbert JP, Raine T, et al. ECCO
Guidelines on Therapeutics in Crohn's Disease: Medical Treatment. Journal of Crohn's and
Colitis. 2019;14(1):4-22.

82
104. Rezaie A, Kuenzig ME, Benchimol EI, Griffiths AM, Otley AR, Steinhart AH, et al.
Budesonide for induction of remission in Crohn's disease. Cochrane Database of Systematic
Reviews. 2015(6).
105. Moja L, Danese S, Fiorino G, Del Giovane C, Bonovas S. Systematic review with
network meta-analysis: comparative efficacy and safety of budesonide and mesalazine
(mesalamine) for Crohn's disease. Aliment Pharmacol Ther. 2015;41(11):1055-65.
106. Benchimol EI, Seow CH, Steinhart AH, Griffiths AM. Traditional corticosteroids for
induction of remission in Crohn's disease. Cochrane Database of Systematic Reviews.
2008(2).
107. Torres J, Bonovas S, Doherty G, Kucharzik T, Gisbert JP, Raine T, et al. ECCO
Guidelines on Therapeutics in Crohn's Disease: Medical Treatment. Journal of Crohn's and
Colitis. 2020;14(1):4-22.
108. Stidham R, Lee T, Higgins P, Deshpande A, Sussman D, Singal A, et al. Systematic
review with network meta‐analysis: the efficacy of anti‐TNF agents for the treatment of
Crohn's disease. Alimentary pharmacology & therapeutics. 2014;39(12):1349-62.
109. Cholapranee A, Hazlewood GS, Kaplan GG, Peyrin‐Biroulet L, Ananthakrishnan AN.
Systematic review with meta‐analysis: comparative efficacy of biologics for induction and
maintenance of mucosal healing in Crohn's disease and ulcerative colitis controlled trials.
Alimentary pharmacology & therapeutics. 2017;45(10):1291-302.
110. Ford AC, Achkar J-P, Khan KJ, Kane SV, Talley NJ, Marshall JK, et al. Efficacy of 5-
aminosalicylates in ulcerative colitis: systematic review and meta-analysis. Official journal of
the American College of Gastroenterology| ACG. 2011;106(4):601-16.
111. Hazlewood GS, Rezaie A, Borman M, Panaccione R, Ghosh S, Seow CH, et al.
Comparative effectiveness of immunosuppressants and biologics for inducing and
maintaining remission in Crohn's disease: a network meta-analysis. Gastroenterology.
2015;148(2):344-54. e5.
112. Matsumoto T, Motoya S, Watanabe K, Hisamatsu T, Nakase H, Yoshimura N, et al.
Adalimumab monotherapy and a combination with azathioprine for Crohn’s disease: a
prospective, randomized trial. Journal of Crohn's and Colitis. 2016;10(11):1259-66.
113. Colombel JF, Sandborn WJ, Reinisch W, Mantzaris GJ, Kornbluth A, Rachmilewitz D,
et al. Infliximab, azathioprine, or combination therapy for Crohn's disease. New England
journal of medicine. 2010;362(15):1383-95.
114. Sands BE, Sandborn WJ, Panaccione R, O'Brien CD, Zhang H, Johanns J, et al.
Ustekinumab as Induction and Maintenance Therapy for Ulcerative Colitis. N Engl J Med.
2019;381(13):1201-14.
115. Sandborn WJ, Gasink C, Gao L-L, Blank MA, Johanns J, Guzzo C, et al. Ustekinumab
induction and maintenance therapy in refractory Crohn's disease. New England Journal of
Medicine. 2012;367(16):1519-28.
116. MacDonald JK, Nguyen TM, Khanna R, Timmer A. Anti‐IL‐12/23p40 antibodies for
induction of remission in Crohn's disease. Cochrane Database of Systematic Reviews.
2016(11).
117. Sandborn WJ, Feagan BG, Rutgeerts P, Hanauer S, Colombel J-F, Sands BE, et al.
Vedolizumab as induction and maintenance therapy for Crohn's disease. New England
Journal of Medicine. 2013;369(8):711-21.
118. Feagan BG, Rutgeerts P, Sands BE, Hanauer S, Colombel JF, Sandborn WJ, et al.
Vedolizumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med.
2013;369(8):699-710.

83
119. Feagan BG, Greenberg GR, Wild G, Fedorak RN, Paré P, McDonald JW, et al.
Treatment of active Crohn's disease with MLN0002, a humanized antibody to the α4β7
integrin. Clinical Gastroenterology and Hepatology. 2008;6(12):1370-7.
120. Sands BE, Feagan BG, Rutgeerts P, Colombel J-F, Sandborn WJ, Sy R, et al. Effects of
vedolizumab induction therapy for patients with Crohn’s disease in whom tumor necrosis
factor antagonist treatment failed. Gastroenterology. 2014;147(3):618-27. e3.
121. Kawalec P, Moćko P. An indirect comparison of ustekinumab and vedolizumab in the
therapy of TNF-failure Crohn's disease patients. Journal of comparative effectiveness
research. 2018;7(2):101-11.
122. Present DH, Rutgeerts P, Targan S, Hanauer SB, Mayer L, Van Hogezand R, et al.
Infliximab for the treatment of fistulas in patients with Crohn's disease. New England Journal
of Medicine. 1999;340(18):1398-405.
123. Feroz SH, Ahmed A, Muralidharan A, Thirunavukarasu P. Comparison of the Efficacy
of the Various Treatment Modalities in the Management of Perianal Crohn's Fistula: A
Review. Cureus. 2020;12(12):e11882.
124. Lichtiger S, Binion D, Wolf D, Present D, Bensimon A, Wu E, et al. The CHOICE trial:
adalimumab demonstrates safety, fistula healing, improved quality of life and increased
work productivity in patients with Crohn’s disease who failed prior infliximab therapy.
Alimentary pharmacology & therapeutics. 2010;32(10):1228-39.
125. Panés J, Rimola J. Perianal fistulizing Crohn's disease: pathogenesis, diagnosis and
therapy. Nature reviews Gastroenterology & hepatology. 2017;14(11):652-64.
126. Feagan BG, MacDonald JK. Oral 5‐aminosalicylic acid for induction of remission in
ulcerative colitis. Cochrane database of systematic reviews. 2012(10).
127. Wang Y, Parker CE, Bhanji T, Feagan BG, MacDonald JK. Oral 5‐aminosalicylic acid for
induction of remission in ulcerative colitis. Cochrane Database of Systematic Reviews.
2016(4).
128. D'Haens G, Kovacs A, Vergauwe P, Nagy F, Molnar T, Bouhnik Y, et al. Clinical trial:
Preliminary efficacy and safety study of a new Budesonide-MMX® 9 mg extended-release
tablets in patients with active left-sided ulcerative colitis. Journal of Crohn's and Colitis.
2010;4(2):153-60.
129. Sandborn WJ, Travis S, Moro L, Jones R, Gautille T, Bagin R, et al. Once-daily
budesonide MMX® extended-release tablets induce remission in patients with mild to
moderate ulcerative colitis: results from the CORE I study. Gastroenterology.
2012;143(5):1218-26. e2.
130. Travis SP, Danese S, Kupcinskas L, Alexeeva O, D'Haens G, Gibson PR, et al. Once-
daily budesonide MMX in active, mild-to-moderate ulcerative colitis: results from the
randomised CORE II study. Gut. 2014;63(3):433-41.
131. Rubin D, Cohen R, Sandborn W, Lichtenstein G, Axler J, Riddell R, et al. Budesonide
MMX® 9 mg for inducing remission in patients with mild-to-moderate ulcerative colitis not
adequately controlled with oral 5-ASAs. J Crohns Colitis. 2015;9(Suppl 1):S7.
132. Danese S, Siegel C, Peyrin‐Biroulet L. integrating budesonide‐MMX into treatment
algorithms for mild‐to‐moderate ulcerative colitis. Alimentary pharmacology & therapeutics.
2014;39(10):1095-103.
133. Löfberg R, Danielsson Å, Suhr O, Nilsson Å, Schiöler R, Nyberg A, et al. Oral
budesonide versus prednisolone in patients with extensive and left sided ulcerative colitis.
Gastroenterology. 1996;110(6):1713-8.

84
134. Khan KJ, Dubinsky MC, Ford AC, Ullman TA, Talley NJ, Moayyedi P. Efficacy of
immunosuppressive therapy for inflammatory bowel disease: a systematic review and meta-
analysis. Official journal of the American College of Gastroenterology| ACG.
2011;106(4):630-42.
135. Gisbert JP, Linares PM, McNicholl AG, Maté J, Gomollón F. Meta-analysis: the
efficacy of azathioprine and mercaptopurine in ulcerative colitis. Aliment Pharmacol Ther.
2009;30(2):126-37.
136. Timmer A, Patton PH, Chande N, McDonald JW, MacDonald JK. Azathioprine and 6-
mercaptopurine for maintenance of remission in ulcerative colitis. Cochrane Database Syst
Rev. 2016;2016(5):Cd000478.
137. Sands B, Peyrin-Biroulet L, Loftus E, Danese S, Colombel J, Abhyankar B, et al. 416a –
Vedolizumab Shows Superior Efficacy Versus Adalimumab: Results of Varsity—The First
Head-To-Head Study of Biologic Therapy for Moderate-To-Severe Ulcerative Colitis.
Gastroenterology. 2019;156:S-81.
138. Sardesai A, Dignass A, Quon P, Milev S, Cappelleri JC, Kisser A, et al. Cost-
effectiveness of tofacitinib compared with infliximab, adalimumab, golimumab, vedolizumab
and ustekinumab for the treatment of moderate to severe ulcerative colitis in Germany. J
Med Econ. 2021;24(1):279-90.
139. Sandborn W, Sands B, Panaccione R, O’Brien C, Zhang H, Johanns J, et al. OP37
Efficacy and safety of ustekinumab as maintenance therapy in ulcerative colitis: Week 44
results from UNIFI. Journal of Crohn's and Colitis. 2019;13(Supplement_1):S025-S6.
140. Singh S, Murad MH, Fumery M, Dulai PS, Sandborn WJ. First- and Second-Line
Pharmacotherapies for Patients With Moderate to Severely Active Ulcerative Colitis: An
Updated Network Meta-Analysis. Clinical gastroenterology and hepatology : the official
clinical practice journal of the American Gastroenterological Association. 2020;18(10):2179-
91.e6.
141. gov C. Safety study of tofacitinib versus tumor necrosis factor (TNF) inhibitor in
subjects with rheumatoid arthritis. 2014.
142. Wang Y, MacDonald JK, Vandermeer B, Griffiths AM, El‐Matary W. Methotrexate for
maintenance of remission in ulcerative colitis. Cochrane Database of Systematic Reviews.
2015(8).
143. Carbonnel F, Colombel JF, Filippi J, Katsanos KH, Peyrin-Biroulet L, Allez M, et al.
Methotrexate is not superior to placebo for inducing steroid-free remission, but induces
steroid-free clinical remission in a larger proportion of patients with ulcerative colitis.
Gastroenterology. 2016;150(2):380-8. e4.
144. Panaccione R, Ghosh S, Middleton S, Márquez JR, Scott BB, Flint L, et al. Combination
therapy with infliximab and azathioprine is superior to monotherapy with either agent in
ulcerative colitis. Gastroenterology. 2014;146(2):392-400. e3.
145. Singh S, Allegretti JR, Siddique SM, Terdiman JP. AGA Technical Review on the
Management of Moderate to Severe Ulcerative Colitis. Gastroenterology. 2020;158(5):1465-
96.e17.
146. Chapman TP, Frias Gomes C, Louis E, Colombel JF, Satsangi J. Review article:
withdrawal of 5-aminosalicylates in inflammatory bowel disease. Aliment Pharmacol Ther.
2020;52(1):73-84.
147. Doherty G, Katsanos KH, Burisch J, Allez M, Papamichael K, Stallmach A, et al.
European Crohn's and Colitis Organisation Topical Review on Treatment Withdrawal ['Exit
Strategies'] in Inflammatory Bowel Disease. J Crohns Colitis. 2018;12(1):17-31.

85
148. Na SY, Moon W. Perspectives on Current and Novel Treatments for Inflammatory
Bowel Disease. Gut Liver. 2019;13(6):604-16.
149. Shen B, Kochhar G, Navaneethan U, Liu X, Farraye FA, Gonzalez-Lama Y, et al. Role of
interventional inflammatory bowel disease in the era of biologic therapy: a position
statement from the Global Interventional IBD Group. Gastrointest Endosc. 2019;89(2):215-
37.
150. Bhattacharya A, Osterman MT. Biologic Therapy for Ulcerative Colitis. Gastroenterol
Clin North Am. 2020;49(4):717-29.
151. Sandborn WJ, Schreiber S, Hanauer SB, Colombel JF, Bloomfield R, Lichtenstein GR.
Reinduction with certolizumab pegol in patients with relapsed Crohn's disease: results from
the PRECiSE 4 Study. Clin Gastroenterol Hepatol. 2010;8(8):696-702.e1.
152. Lichtenstein G, Thomsen OO, Schreiber S, Lawrance IC, Hanauer SB, Bloomfield R, et
al. S1040 Long-Term Remission With Certolizumab Pegol in Crohn's Disease: Efficacy Over 4
Years in Patients With NO Prior TNF-α Inhibitor Exposure (PRECiSE 3 Study).
Gastroenterology. 2010;5(138):S-165.
153. Schreiber S, Colombel JF, Bloomfield R, Nikolaus S, Schölmerich J, Panés J, et al.
Increased response and remission rates in short-duration Crohn's disease with
subcutaneous certolizumab pegol: an analysis of PRECiSE 2 randomized maintenance trial
data. Am J Gastroenterol. 2010;105(7):1574-82.
154. Sandborn WJ, Feagan BG, Stoinov S, Honiball PJ, Rutgeerts P, Mason D, et al.
Certolizumab pegol for the treatment of Crohn's disease. N Engl J Med. 2007;357(3):228-38.
155. Berg DR, Colombel JF, Ungaro R. The Role of Early Biologic Therapy in Inflammatory
Bowel Disease. Inflamm Bowel Dis. 2019;25(12):1896-905.
156. Feuerstein JD, Isaacs KL, Schneider Y, Siddique SM, Falck-Ytter Y, Singh S. AGA Clinical
Practice Guidelines on the Management of Moderate to Severe Ulcerative Colitis.
Gastroenterology. 2020;158(5):1450-61.
157. Pabla BS, Schwartz DA. Assessing Severity of Disease in Patients with Ulcerative
Colitis. Gastroenterol Clin North Am. 2020;49(4):671-88.
158. D'Haens G, Baert F, van Assche G, Caenepeel P, Vergauwe P, Tuynman H, et al. Early
combined immunosuppression or conventional management in patients with newly
diagnosed Crohn's disease: an open randomised trial. Lancet. 2008;371(9613):660-7.
159. Nielsen OH, Steenholdt C, Juhl CB, Rogler G. Efficacy and safety of methotrexate in
the management of inflammatory bowel disease: A systematic review and meta-analysis of
randomized, controlled trials. EClinicalMedicine. 2020;20.
160. Lichtenstein GR, Loftus EV, Isaacs KL, Regueiro MD, Gerson LB, Sands BE. ACG Clinical
Guideline: Management of Crohn's Disease in Adults. Am J Gastroenterol. 2018;113(4):481-
517.
161. Cottone M, Kohn A, Daperno M, Armuzzi A, Guidi L, D'Inca R, et al. Advanced age is
an independent risk factor for severe infections and mortality in patients given anti-tumor
necrosis factor therapy for inflammatory bowel disease. Clin Gastroenterol Hepatol.
2011;9(1):30-5.
162. Toruner M, Loftus EV, Jr., Harmsen WS, Zinsmeister AR, Orenstein R, Sandborn WJ,
et al. Risk factors for opportunistic infections in patients with inflammatory bowel disease.
Gastroenterology. 2008;134(4):929-36.
163. Peyrin-Biroulet L, Khosrotehrani K, Carrat F, Bouvier AM, Chevaux JB, Simon T, et al.
Increased risk for nonmelanoma skin cancers in patients who receive thiopurines for
inflammatory bowel disease. Gastroenterology. 2011;141(5):1621-28.e1-5.

86
164. Yajnik V, Khan N, Dubinsky M, Axler J, James A, Abhyankar B, et al. Efficacy and
Safety of Vedolizumab in Ulcerative Colitis and Crohn's Disease Patients Stratified by Age.
Adv Ther. 2017;34(2):542-59.
165. Bonovas S, Fiorino G, Allocca M, Lytras T, Nikolopoulos GK, Peyrin-Biroulet L, et al.
Biologic Therapies and Risk of Infection and Malignancy in Patients With Inflammatory
Bowel Disease: A Systematic Review and Network Meta-analysis. Clin Gastroenterol
Hepatol. 2016;14(10):1385-97.e10.
166. Feagan BG, Schwartz D, Danese S, Rubin DT, Lissoos TW, Xu J, et al. Efficacy of
Vedolizumab in Fistulising Crohn's Disease: Exploratory Analyses of Data from GEMINI 2. J
Crohns Colitis. 2018;12(5):621-6.
167. Perry C, Fischer K, Elmoursi A, Kern C, Currier A, Kudaravalli P, et al. Vedolizumab
Dose Escalation Improves Therapeutic Response in a Subset of Patients with Ulcerative
Colitis. Dig Dis Sci. 2021;66(6):2051-8.
168. Samaan MA, Birdi S, Morales MS, Honap S, Tamilarasan AG, Cunningham G, et al.
Effectiveness of vedolizumab dose intensification to achieve inflammatory bowel disease
control in cases of suboptimal response. Frontline Gastroenterol. 2020;11(3):188-93.
169. Ollech JE, Normatov I, Peleg N, Wang J, Patel SA, Rai V, et al. Effectiveness of
Ustekinumab Dose Escalation in Patients With Crohn's Disease. Clin Gastroenterol Hepatol.
2021;19(1):104-10.
170. Haider SA, Yadav A, Perry C, Su L, Akanbi O, Kudaravalli P, et al. Ustekinumab dose
escalation improves clinical responses in refractory Crohn's disease. Therap Adv
Gastroenterol. 2020;13:1756284820959245.
171. Kopylov U, Hanzel J, Liefferinckx C, De Marco D, Imperatore N, Plevris N, et al.
Effectiveness of ustekinumab dose escalation in Crohn's disease patients with insufficient
response to standard-dose subcutaneous maintenance therapy. Aliment Pharmacol Ther.
2020;52(1):135-42.
172. Mitrev N, Vande Casteele N, Seow CH, Andrews JM, Connor SJ, Moore GT, et al.
Review article: consensus statements on therapeutic drug monitoring of anti-tumour
necrosis factor therapy in inflammatory bowel diseases. Aliment Pharmacol Ther.
2017;46(11-12):1037-53.
173. Ben-Horin S, Chowers Y. Review article: loss of response to anti-TNF treatments in
Crohn's disease. Aliment Pharmacol Ther. 2011;33(9):987-95.
174. Fine S, Papamichael K, Cheifetz AS. Etiology and Management of Lack or Loss of
Response to Anti-Tumor Necrosis Factor Therapy in Patients With Inflammatory Bowel
Disease. Gastroenterol Hepatol (N Y). 2019;15(12):656-65.
175. Feagan BG, Rubin DT, Danese S, Vermeire S, Abhyankar B, Sankoh S, et al. Efficacy of
Vedolizumab Induction and Maintenance Therapy in Patients With Ulcerative Colitis,
Regardless of Prior Exposure to Tumor Necrosis Factor Antagonists. Clin Gastroenterol
Hepatol. 2017;15(2):229-39.e5.
176. Carrascosa JM. Immunogenicity in biologic therapy: implications for dermatology.
Actas Dermosifiliogr. 2013;104(6):471-9.
177. Grossi V, Gulli F, Infantino M, Stefanile A, Napodano C, Benucci M, et al. The
Laboratory Role in anti-TNF Biological Therapy Era. Immunol Invest. 2020;49(3):317-32.
178. Plasencia C, Pascual-Salcedo D, García-Carazo S, Lojo L, Nuño L, Villalba A, et al. The
immunogenicity to the first anti-TNF therapy determines the outcome of switching to a
second anti-TNF therapy in spondyloarthritis patients. Arthritis Res Ther. 2013;15(4):R79.

87
179. Gisbert JP, Marín AC, McNicholl AG, Chaparro M. Systematic review with meta-
analysis: the efficacy of a second anti-TNF in patients with inflammatory bowel disease
whose previous anti-TNF treatment has failed. Aliment Pharmacol Ther. 2015;41(7):613-23.
180. Kothari MM, Nguyen DL, Parekh NK. Strategies for overcoming anti-tumor necrosis
factor drug antibodies in inflammatory bowel disease: Case series and review of literature.
World J Gastrointest Pharmacol Ther. 2017;8(3):155-61.
181. Schräpel C, Kovar L, Selzer D, Hofmann U, Tran F, Reinisch W, et al. External Model
Performance Evaluation of Twelve Infliximab Population Pharmacokinetic Models in Patients
with Inflammatory Bowel Disease. Pharmaceutics. 2021;13(9):1368.
182. Roblin X, Marotte H, Rinaudo M, Del Tedesco E, Moreau A, Phelip JM, et al.
Association between pharmacokinetics of adalimumab and mucosal healing in patients with
inflammatory bowel diseases. Clin Gastroenterol Hepatol. 2014;12(1):80-4.e2.
183. Bush MC, Patel S, Lapinski RH, Stone JL. Perinatal outcomes in inflammatory bowel
disease. J Matern Fetal Neonatal Med. 2004;15(4):237-41.
184. Morales M, Berney T, Jenny A, Morel P, Extermann P. Crohn's disease as a risk factor
for the outcome of pregnancy. Hepatogastroenterology. 2000;47(36):1595-8.
185. Magro F, Gionchetti P, Eliakim R, Ardizzone S, Armuzzi A, Barreiro-de Acosta M, et al.
Third European Evidence-based Consensus on Diagnosis and Management of Ulcerative
Colitis. Part 1: Definitions, Diagnosis, Extra-intestinal Manifestations, Pregnancy, Cancer
Surveillance, Surgery, and Ileo-anal Pouch Disorders. J Crohns Colitis. 2017;11(6):649-70.
186. Zelinkova Z, van der Ent C, Bruin KF, van Baalen O, Vermeulen HG, Smalbraak HJ, et
al. Effects of discontinuing anti-tumor necrosis factor therapy during pregnancy on the
course of inflammatory bowel disease and neonatal exposure. Clin Gastroenterol Hepatol.
2013;11(3):318-21.
187. de Lima A, Zelinkova Z, van der Ent C, Steegers EA, van der Woude CJ. Tailored anti-
TNF therapy during pregnancy in patients with IBD: maternal and fetal safety. Gut.
2016;65(8):1261-8.
188. Beaulieu DB, Ananthakrishnan AN, Martin C, Cohen RD, Kane SV, Mahadevan U. Use
of Biologic Therapy by Pregnant Women With Inflammatory Bowel Disease Does Not Affect
Infant Response to Vaccines. Clin Gastroenterol Hepatol. 2018;16(1):99-105.
189. Papp KA, Haraoui B, Kumar D, Marshall JK, Bissonnette R, Bitton A, et al. Vaccination
Guidelines for Patients With Immune-Mediated Disorders on Immunosuppressive Therapies.
J Cutan Med Surg. 2019;23(1):50-74.
190. Park SH, Kim HJ, Lee CK, Song EM, Kang S-B, Jang BI, et al. Safety and Optimal Timing
of BCG Vaccination in Infants Born to Mothers Receiving Anti-TNF Therapy for Inflammatory
Bowel Disease. Journal of Crohn's and Colitis. 2020;14(12):1780-4.
191. Heller MM, Wu JJ, Murase JE. Fatal case of disseminated BCG infection after
vaccination of an infant with in utero exposure to infliximab. Journal of the American
Academy of Dermatology. 2011;65(4):870.
192. Bendaoud S, Nahon S, Gornet J-M, Pariente B, Beaugerie L, Abitbol V, et al. P817
Live-vaccines and lactation in newborn exposed in utero to anti-TNF: A multi-centre French
experience in inflammatory bowel disease. Journal of Crohn's and Colitis.
2018;12(supplement_1):S527-S.
193. Trallori G, d'Albasio G, Bardazzi G, Bonanomi AG, Amorosi A, Del Carlo P, et al. 5-
Aminosalicylic acid in pregnancy: clinical report. Ital J Gastroenterol. 1994;26(2):75-8.

88
194. De Broe ME, Stolear JC, Nouwen EJ, Elseviers MM. 5-Aminosalicylic acid (5-ASA) and
chronic tubulointerstitial nephritis in patients with chronic inflammatory bowel disease: is
there a link? Nephrol Dial Transplant. 1997;12(9):1839-41.
195. Bell CM, Habal FM. Safety of topical 5-aminosalicylic acid in pregnancy. Am J
Gastroenterol. 1997;92(12):2201-2.
196. Marteau P, Tennenbaum R, Elefant E, Lémann M, Cosnes J. Foetal outcome in
women with inflammatory bowel disease treated during pregnancy with oral mesalazine
microgranules. Aliment Pharmacol Ther. 1998;12(11):1101-8.
197. Goldstein LH, Dolinsky G, Greenberg R, Schaefer C, Cohen-Kerem R, Diav-Citrin O, et
al. Pregnancy outcome of women exposed to azathioprine during pregnancy. Birth Defects
Res A Clin Mol Teratol. 2007;79(10):696-701.
198. Mahadevan U, Wolf DC, Dubinsky M, Cortot A, Lee SD, Siegel CA, et al. Placental
transfer of anti-tumor necrosis factor agents in pregnant patients with inflammatory bowel
disease. Clin Gastroenterol Hepatol. 2013;11(3):286-92; quiz e24.
199. Nguyen GC, Seow CH, Maxwell C, Huang V, Leung Y, Jones J, et al. The Toronto
Consensus Statements for the Management of Inflammatory Bowel Disease in Pregnancy.
Gastroenterology. 2016;150(3):734-57.e1.
200. Kawaguchi R, Haruta S, Kobayashi H. Efficacy and safety of venous thromboembolism
prophylaxis with fondaparinux in women at risk after cesarean section. Obstet Gynecol Sci.
2017;60(6):535-41.
201. Bar-Gil Shitrit A, Grisaru-Granovsky S, Ben Ya'acov A, Goldin E. Management of
Inflammatory Bowel Disease During Pregnancy. Dig Dis Sci. 2016;61(8):2194-204.
202. Matro R, Martin CF, Wolf D, Shah SA, Mahadevan U. Exposure Concentrations of
Infants Breastfed by Women Receiving Biologic Therapies for Inflammatory Bowel Diseases
and Effects of Breastfeeding on Infections and Development. Gastroenterology.
2018;155(3):696-704.
203. Kanis SL, van der Woude CJ. Proper Use of Inflammatory Bowel Disease Drugs during
Pregnancy. Digestive Diseases. 2016;34(suppl 1)(Suppl. 1):61-6.
204. Saarikoski S, Seppälä M. Immunosuppression during pregnancy: transmission of
azathioprine and its metabolites from the mother to the fetus. Am J Obstet Gynecol.
1973;115(8):1100-6.
205. Casanova MJ, Chaparro M, Domènech E, Barreiro-de Acosta M, Bermejo F, Iglesias E,
et al. Safety of thiopurines and anti-TNF-α drugs during pregnancy in patients with
inflammatory bowel disease. Am J Gastroenterol. 2013;108(3):433-40.
206. Coelho J, Beaugerie L, Colombel JF, Hébuterne X, Lerebours E, Lémann M, et al.
Pregnancy outcome in patients with inflammatory bowel disease treated with thiopurines:
cohort from the CESAME Study. Gut. 2011;60(2):198-203.
207. Zelinkova Z, de Haar C, de Ridder L, Pierik MJ, Kuipers EJ, Peppelenbosch MP, et al.
High intra-uterine exposure to infliximab following maternal anti-TNF treatment during
pregnancy. Aliment Pharmacol Ther. 2011;33(9):1053-8.
208. Narula N, Al-Dabbagh R, Dhillon A, Sands BE, Marshall JK. Anti-TNFα therapies are
safe during pregnancy in women with inflammatory bowel disease: a systematic review and
meta-analysis. Inflamm Bowel Dis. 2014;20(10):1862-9.
209. Lamb CA, Kennedy NA, Raine T, Hendy PA, Smith PJ, Limdi JK, et al. British Society of
Gastroenterology consensus guidelines on the management of inflammatory bowel disease
in adults. Gut. 2019;68(Suppl 3):s1-s106.

89
210. Temprano KK, Bandlamudi R, Moore TL. Antirheumatic drugs in pregnancy and
lactation. Semin Arthritis Rheum. 2005;35(2):112-21.
211. Banaszkiewicz A, Kądzielska J, Gawrońska A, Pituch H, Obuch-Woszczatyński P,
Albrecht P, et al. Enterotoxigenic Clostridium perfringens infection and pediatric patients
with inflammatory bowel disease. J Crohns Colitis. 2014;8(4):276-81.
212. Hourigan SK, Sears CL, Oliva-Hemker M. Clostridium difficile Infection in Pediatric
Inflammatory Bowel Disease. Inflamm Bowel Dis. 2016;22(4):1020-5.
213. Mack DR, Langton C, Markowitz J, LeLeiko N, Griffiths A, Bousvaros A, et al.
Laboratory values for children with newly diagnosed inflammatory bowel disease.
Pediatrics. 2007;119(6):1113-9.
214. Trivedi PJ, Chapman RW. PSC, AIH and overlap syndrome in inflammatory bowel
disease. Clin Res Hepatol Gastroenterol. 2012;36(5):420-36.
215. Sidler MA, Leach ST, Day AS. Fecal S100A12 and fecal calprotectin as noninvasive
markers for inflammatory bowel disease in children. Inflamm Bowel Dis. 2008;14(3):359-66.
216. Sipponen T, Savilahti E, Kolho KL, Nuutinen H, Turunen U, Färkkilä M. Crohn's disease
activity assessed by fecal calprotectin and lactoferrin: correlation with Crohn's disease
activity index and endoscopic findings. Inflamm Bowel Dis. 2008;14(1):40-6.
217. Cabrera-Abreu JC, Davies P, Matek Z, Murphy MS. Performance of blood tests in
diagnosis of inflammatory bowel disease in a specialist clinic. Arch Dis Child. 2004;89(1):69-
71.
218. Henderson P, Casey A, Lawrence SJ, Kennedy NA, Kingstone K, Rogers P, et al. The
diagnostic accuracy of fecal calprotectin during the investigation of suspected pediatric
inflammatory bowel disease. Am J Gastroenterol. 2012;107(6):941-9.
219. de Bie CI, Buderus S, Sandhu BK, de Ridder L, Paerregaard A, Veres G, et al.
Diagnostic workup of paediatric patients with inflammatory bowel disease in Europe: results
of a 5-year audit of the EUROKIDS registry. J Pediatr Gastroenterol Nutr. 2012;54(3):374-80.
220. Levine A, Koletzko S, Turner D, Escher JC, Cucchiara S, de Ridder L, et al. ESPGHAN
revised porto criteria for the diagnosis of inflammatory bowel disease in children and
adolescents. J Pediatr Gastroenterol Nutr. 2014;58(6):795-806.
221. Tully MA. Pediatric celiac disease. Gastroenterol Nurs. 2008;31(2):132-40; quiz 41-2.
222. Giles E, Barclay AR, Chippington S, Wilson DC. Systematic review: MRI enterography
for assessment of small bowel involvement in paediatric Crohn's disease. Aliment Pharmacol
Ther. 2013;37(12):1121-31.
223. Essary B, Kim J, Anupindi S, Katz JA, Nimkin K. Pelvic MRI in children with Crohn
disease and suspected perianal involvement. Pediatr Radiol. 2007;37(2):201-8.
224. Liao Z, Gao R, Xu C, Li ZS. Indications and detection, completion, and retention rates
of small-bowel capsule endoscopy: a systematic review. Gastrointest Endosc.
2010;71(2):280-6.
225. Cohen SA, Klevens AI. Use of capsule endoscopy in diagnosis and management of
pediatric patients, based on meta-analysis. Clin Gastroenterol Hepatol. 2011;9(6):490-6.
226. Cheifetz AS, Kornbluth AA, Legnani P, Schmelkin I, Brown A, Lichtiger S, 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):2218-22.
227. Fritscher-Ravens A, Scherbakov P, Bufler P, Torroni F, Ruuska T, Nuutinen H, et al.
The feasibility of wireless capsule endoscopy in detecting small intestinal pathology in
children under the age of 8 years: a multicentre European study. Gut. 2009;58(11):1467-72.

90
228. Chiorean L, Schreiber-Dietrich D, Braden B, Cui X-W, Buchhorn R, Chang J-M, et al.
Ultrasonographic imaging of inflammatory bowel disease in pediatric patients. World J
Gastroenterol. 2015;21(17):5231-41.
229. van Wassenaer EA, Benninga MA, van Limbergen JL, D’Haens GR, Griffiths AM, Koot
BGP. Intestinal Ultrasound in Pediatric Inflammatory Bowel Disease: Promising, but Work in
Progress. Inflammatory Bowel Diseases. 2021.
230. Bremner AR, Griffiths M, Argent JD, Fairhurst JJ, Beattie RM. Sonographic evaluation
of inflammatory bowel disease: a prospective, blinded, comparative study. Pediatr Radiol.
2006;36(9):947-53.
231. Uhlig HH, Schwerd T, Koletzko S, Shah N, Kammermeier J, Elkadri A, et al. The
diagnostic approach to monogenic very early onset inflammatory bowel disease.
Gastroenterology. 2014;147(5):990-1007.e3.
232. Kelsen JR, Baldassano RN, Artis D, Sonnenberg GF. Maintaining intestinal health: the
genetics and immunology of very early onset inflammatory bowel disease. Cell Mol
Gastroenterol Hepatol. 2015;1(5):462-76.
233. Ouahed J, Spencer E, Kotlarz D, Shouval DS, Kowalik M, Peng K, et al. Very Early
Onset Inflammatory Bowel Disease: A Clinical Approach With a Focus on the Role of
Genetics and Underlying Immune Deficiencies. Inflamm Bowel Dis. 2020;26(6):820-42.
234. Turner D, Ruemmele FM, Orlanski-Meyer E, Griffiths AM, de Carpi JM, Bronsky J, et
al. Management of Paediatric Ulcerative Colitis, Part 1: Ambulatory Care-An Evidence-based
Guideline From European Crohn's and Colitis Organization and European Society of
Paediatric Gastroenterology, Hepatology and Nutrition. J Pediatr Gastroenterol Nutr.
2018;67(2):257-91.
235. Annese V, Daperno M, Rutter MD, Amiot A, Bossuyt P, East J, et al. European
evidence based consensus for endoscopy in inflammatory bowel disease. Journal of Crohn's
and Colitis. 2013;7(12):982-1018.
236. Thakkar K, Lucia CJ, Ferry GD, McDuffie A, Watson K, Tsou M, et al. Repeat
endoscopy affects patient management in pediatric inflammatory bowel disease. Am J
Gastroenterol. 2009;104(3):722-7.
237. Kayton ML. Cancer and pediatric inflammatory bowel disease. Semin Pediatr Surg.
2007;16(3):205-13.
238. Waugh N, Cummins E, Royle P, Kandala NB, Shyangdan D, Arasaradnam R, et al.
Faecal calprotectin testing for differentiating amongst inflammatory and non-inflammatory
bowel diseases: systematic review and economic evaluation. Health Technol Assess.
2013;17(55):xv-xix, 1-211.
239. Murray A, Nguyen TM, Parker CE, Feagan BG, MacDonald JK. Oral 5-aminosalicylic
acid for induction of remission in ulcerative colitis. Cochrane Database Syst Rev.
2020;8(8):Cd000543.
240. Winter HS, Krzeski P, Heyman MB, Ibarguen-Secchia E, Iwanczak B, Kaczmarski M, et
al. High- and low-dose oral delayed-release mesalamine in children with mild-to-moderately
active ulcerative colitis. J Pediatr Gastroenterol Nutr. 2014;59(6):767-72.
241. Turner D, Yerushalmi B, Kori M, Broide E, Mozer-Glassberg Y, Shaoul R, et al. Once-
Versus Twice-daily Mesalazine to Induce Remission in Paediatric Ulcerative Colitis: A
Randomised Controlled Trial. J Crohns Colitis. 2017;11(5):527-33.
242. Quiros JA, Heyman MB, Pohl JF, Attard TM, Pieniaszek HJ, Bortey E, et al. Safety,
efficacy, and pharmacokinetics of balsalazide in pediatric patients with mild-to-moderate

91
active ulcerative colitis: results of a randomized, double-blind study. J Pediatr Gastroenterol
Nutr. 2009;49(5):571-9.
243. Probert CS, Dignass AU, Lindgren S, Oudkerk Pool M, Marteau P. Combined oral and
rectal mesalazine for the treatment of mild-to-moderately active ulcerative colitis: rapid
symptom resolution and improvements in quality of life. J Crohns Colitis. 2014;8(3):200-7.
244. Marteau P, Probert CS, Lindgren S, Gassul M, Tan TG, Dignass A, et al. Combined oral
and enema treatment with Pentasa (mesalazine) is superior to oral therapy alone in patients
with extensive mild/moderate active ulcerative colitis: a randomised, double blind, placebo
controlled study. Gut. 2005;54(7):960-5.
245. Heyman MB, Kierkus J, Spénard J, Shbaklo H, Giguere M. Efficacy and safety of
mesalamine suppositories for treatment of ulcerative proctitis in children and adolescents.
Inflamm Bowel Dis. 2010;16(11):1931-9.
246. Tung J, Loftus EV, Jr., Freese DK, El-Youssef M, Zinsmeister AR, Melton LJ, 3rd, et al. A
population-based study of the frequency of corticosteroid resistance and dependence in
pediatric patients with Crohn's disease and ulcerative colitis. Inflamm Bowel Dis.
2006;12(12):1093-100.
247. Cakir M, Ozgenc F, Yusekkaya HA, Ecevit CO, Yagci RV. Steroid response in moderate
to severe pediatric ulcerative colitis: a single center's experience. World J Pediatr.
2011;7(1):50-3.
248. Beattie RM, Nicholls SW, Domizio P, Williams CB, Walker-Smith JA. Endoscopic
assessment of the colonic response to corticosteroids in children with ulcerative colitis. J
Pediatr Gastroenterol Nutr. 1996;22(4):373-9.
249. Campieri M, Adamo S, Valpiani D, D'Arienzo A, D'Albasio G, Pitzalis M, et al. Oral
beclometasone dipropionate in the treatment of extensive and left-sided active ulcerative
colitis: a multicentre randomised study. Aliment Pharmacol Ther. 2003;17(12):1471-80.
250. Romano C, Famiani A, Comito D, Rossi P, Raffa V, Fries W. Oral beclomethasone
dipropionate in pediatric active ulcerative colitis: a comparison trial with mesalazine. J
Pediatr Gastroenterol Nutr. 2010;50(4):385-9.
251. Karolewska-Bochenek K, Dziekiewicz M, Banaszkiewicz A. Budesonide MMX in
Paediatric Patients With Ulcerative Colitis. J Crohns Colitis. 2017;11(11):1402.
252. Jakobsen C, Bartek J, Jr., Wewer V, Vind I, Munkholm P, Groen R, et al. Differences in
phenotype and disease course in adult and paediatric inflammatory bowel disease--a
population-based study. Aliment Pharmacol Ther. 2011;34(10):1217-24.
253. Hyams JS, Lerer T, Mack D, Bousvaros A, Griffiths A, Rosh J, et al. Outcome following
thiopurine use in children with ulcerative colitis: a prospective multicenter registry study.
Am J Gastroenterol. 2011;106(5):981-7.
254. Aloi M, DʼArcangelo G, Bramuzzo M, Gasparetto M, Martinelli M, Alvisi P, et al. Effect
of Early Versus Late Azathioprine Therapy in Pediatric Ulcerative Colitis. Inflamm Bowel Dis.
2016;22(7):1647-54.
255. Wang Y, Parker CE, Feagan BG, MacDonald JK. Oral 5-aminosalicylic acid for
maintenance of remission in ulcerative colitis. Cochrane Database Syst Rev.
2016;2016(5):Cd000544.
256. Tajiri H, Tomomasa T, Yoden A, Konno M, Sasaki M, Maisawa S, et al. Efficacy and
safety of azathioprine and 6-mercaptopurine in Japanese pediatric patients with ulcerative
colitis: a survey of the Japanese Society for Pediatric Inflammatory Bowel Disease. Digestion.
2008;77(3-4):150-4.

92
257. Pozler O, Chládek J, Malý J, Hroch M, Dědek P, Beránek M, et al. Steady-state of
azathioprine during initiation treatment of pediatric inflammatory bowel disease. J Crohns
Colitis. 2010;4(6):623-8.
258. Timmer A, McDonald JW, Macdonald JK. Azathioprine and 6-mercaptopurine for
maintenance of remission in ulcerative colitis. Cochrane Database Syst Rev.
2007(1):Cd000478.
259. Gilissen LPL, Wong DR, Engels LGJB, Bierau J, Bakker JA, Paulussen ADC, et al.
Therapeutic drug monitoring of thiopurine metabolites in adult thiopurine tolerant IBD
patients on maintenance therapy. Journal of Crohn's and Colitis. 2012;6(6):698-707.
260. Lee JH, Kim TJ, Kim ER, Hong SN, Chang DK, Choi L-H, et al. Measurements of 6-
thioguanine nucleotide levels with TPMT and NUDT15 genotyping in patients with Crohn's
disease. PLoS One. 2017;12(12):e0188925-e.
261. Sparrow MP. Use of allopurinol to optimize thiopurine immunomodulator efficacy in
inflammatory bowel disease. Gastroenterol Hepatol (N Y). 2008;4(7):505-11.
262. Hyams J, Damaraju L, Blank M, Johanns J, Guzzo C, Winter HS, et al. Induction and
maintenance therapy with infliximab for children with moderate to severe ulcerative colitis.
Clin Gastroenterol Hepatol. 2012;10(4):391-9.e1.
263. Turner D, Griffiths AM. Acute severe ulcerative colitis in children: a systematic
review. Inflamm Bowel Dis. 2011;17(1):440-9.
264. Thorlund K, Druyts E, Mills EJ, Fedorak RN, Marshall JK. Adalimumab versus
infliximab for the treatment of moderate to severe ulcerative colitis in adult patients naïve
to anti-TNF therapy: an indirect treatment comparison meta-analysis. J Crohns Colitis.
2014;8(7):571-81.
265. Vahabnezhad E, Rabizadeh S, Dubinsky MC. A 10-year, single tertiary care center
experience on the durability of infliximab in pediatric inflammatory bowel disease. Inflamm
Bowel Dis. 2014;20(4):606-13.
266. Volonaki E, Mutalib M, Kiparissi F, Shah N, Lindley KJ, Elawad M. Adalimumab as a
second-line biological therapy in children with refractory ulcerative colitis. Eur J
Gastroenterol Hepatol. 2015;27(12):1425-8.
267. Hyams J, Griffiths A, Veereman G, Turner D, Chan D, Adedokun O, et al. P-097 A
Multicenter Open-Label Study Assessing Pharmacokinetics, Efficacy, and Safety of
Subcutaneous Golimumab in Pediatric Subjects with Moderately-Severely Active Ulcerative
Colitis. Inflammatory Bowel Diseases. 2016;22:S39-S40.
268. Rundquist S, Sachs MC, Eriksson C, Olén O, Montgomery S, Halfvarson J, et al. Drug
survival of anti-TNF agents compared with vedolizumab as a second-line biological
treatment in inflammatory bowel disease: results from nationwide Swedish registers.
Alimentary Pharmacology & Therapeutics. 2021;53(4):471-83.
269. Yanai H, Lichtenstein L, Assa A, Mazor Y, Weiss B, Levine A, et al. Levels of drug and
antidrug antibodies are associated with outcome of interventions after loss of response to
infliximab or adalimumab. Clin Gastroenterol Hepatol. 2015;13(3):522-30.e2.
270. Sands BE, Feagan BG, Rutgeerts P, Colombel JF, Sandborn WJ, Sy R, et al. Effects of
vedolizumab induction therapy for patients with Crohn's disease in whom tumor necrosis
factor antagonist treatment failed. Gastroenterology. 2014;147(3):618-27.e3.
271. Afif W, Loftus EV, Jr., Faubion WA, Kane SV, Bruining DH, Hanson KA, et al. Clinical
utility of measuring infliximab and human anti-chimeric antibody concentrations in patients
with inflammatory bowel disease. Am J Gastroenterol. 2010;105(5):1133-9.

93
272. Dalal SR, Cohen RD. What to Do When Biologic Agents Are Not Working in
Inflammatory Bowel Disease Patients. Gastroenterol Hepatol (N Y). 2015;11(10):657-65.
273. Aardoom MA, Veereman G, de Ridder L. A Review on the Use of Anti-TNF in Children
and Adolescents with Inflammatory Bowel Disease. Int J Mol Sci. 2019;20(10):2529.
274. Dohos D, Hanák L, Szakács Z, Kiss S, Párniczky A, Erőss B, et al. Systematic review
with meta-analysis: the effects of immunomodulator or biological withdrawal from mono-
or combination therapy in inflammatory bowel disease. Aliment Pharmacol Ther.
2021;53(2):220-33.
275. Tarnok A, Kiss Z, Kadenczki O, Veres G. Characteristics of biological therapy in
pediatric patients with Crohn's disease. Expert Opin Biol Ther. 2019;19(3):181-96.
276. Cheon JH. Understanding the complications of anti-tumor necrosis factor therapy in
East Asian patients with inflammatory bowel disease. J Gastroenterol Hepatol.
2017;32(4):769-77.
277. Rahier JF, Magro F, Abreu C, Armuzzi A, Ben-Horin S, Chowers Y, et al. Second
European evidence-based consensus on the prevention, diagnosis and management of
opportunistic infections in inflammatory bowel disease. J Crohns Colitis. 2014;8(6):443-68.
278. Ford AC, Kane SV, Khan KJ, Achkar J-P, Talley NJ, Marshall JK, et al. Efficacy of 5-
aminosalicylates in Crohn's disease: systematic review and meta-analysis. Official journal of
the American College of Gastroenterology| ACG. 2011;106(4):617-29.
279. Lim WC, Wang Y, MacDonald JK, Hanauer S. Aminosalicylates for induction of
remission or response in Crohn's disease. Cochrane Database Syst Rev. 2016;7(7):Cd008870.
280. Coward S, Kuenzig ME, Hazlewood G, Clement F, McBrien K, Holmes R, et al.
Comparative Effectiveness of Mesalamine, Sulfasalazine, Corticosteroids, and Budesonide
for the Induction of Remission in Crohn's Disease: A Bayesian Network Meta-analysis.
Inflamm Bowel Dis. 2017;23(3):461-72.
281. Akobeng AK, Zhang D, Gordon M, MacDonald JK. Oral 5-aminosalicylic acid for
maintenance of medically-induced remission in Crohn's disease. Cochrane Database Syst
Rev. 2016;9(9):Cd003715.
282. Cezard JP, Munck A, Mouterde O, Morali A, Lenaerts C, Lachaux A, et al. Prevention
of relapse by mesalazine (Pentasa) in pediatric Crohn's disease: a multicenter, double-blind,
randomized, placebo-controlled trial. Gastroenterol Clin Biol. 2009;33(1 Pt 1):31-40.
283. Faubion WA, Jr., Loftus EV, Jr., Harmsen WS, Zinsmeister AR, Sandborn WJ. The
natural history of corticosteroid therapy for inflammatory bowel disease: a population-
based study. Gastroenterology. 2001;121(2):255-60.
284. O'Sullivan M. Symposium on 'The challenge of translating nutrition research into
public health nutrition'. Session 3: Joint Nutrition Society and Irish Nutrition and Dietetic
Institute Symposium on 'Nutrition and autoimmune disease'. Nutrition in Crohn's disease.
Proc Nutr Soc. 2009;68(2):127-34.
285. Levine A, Weizman Z, Broide E, Shamir R, Shaoul R, Pacht A, et al. A comparison of
budesonide and prednisone for the treatment of active pediatric Crohn disease. J Pediatr
Gastroenterol Nutr. 2003;36(2):248-52.
286. Levine A, Broide E, Stein M, Bujanover Y, Weizman Z, Dinari G, et al. Evaluation of
oral budesonide for treatment of mild and moderate exacerbations of Crohn's disease in
children. J Pediatr. 2002;140(1):75-80.
287. Kuenzig ME, Rezaie A, Seow CH, Otley AR, Steinhart AH, Griffiths AM, et al.
Budesonide for maintenance of remission in Crohn's disease. Cochrane Database Syst Rev.
2014;2014(8):Cd002913.

94
288. Steinhart AH, Ewe K, Griffiths AM, Modigliani R, Thomsen OO. Corticosteroids for
maintenance of remission in Crohn's disease. Cochrane Database Syst Rev.
2003(4):Cd000301.
289. Johnson T, Macdonald S, Hill SM, Thomas A, Murphy MS. Treatment of active
Crohn's disease in children using partial enteral nutrition with liquid formula: a randomised
controlled trial. Gut. 2006;55(3):356-61.
290. Lee D, Baldassano RN, Otley AR, Albenberg L, Griffiths AM, Compher C, et al.
Comparative Effectiveness of Nutritional and Biological Therapy in North American Children
with Active Crohn's Disease. Inflamm Bowel Dis. 2015;21(8):1786-93.
291. Chande N, Tsoulis DJ, MacDonald JK. Azathioprine or 6-mercaptopurine for induction
of remission in Crohn's disease. Cochrane Database Syst Rev. 2013(4):Cd000545.
292. Khan KJ, Dubinsky MC, Ford AC, Ullman TA, Talley NJ, Moayyedi P. Efficacy of
immunosuppressive therapy for inflammatory bowel disease: a systematic review and meta-
analysis. Am J Gastroenterol. 2011;106(4):630-42.
293. Wenzl HH, Primas C, Novacek G, Teml A, Öfferlbauer-Ernst A, Högenauer C, et al.
Withdrawal of long-term maintenance treatment with azathioprine tends to increase
relapse risk in patients with Crohn's disease. Dig Dis Sci. 2015;60(5):1414-23.
294. Markowitz J, Grancher K, Kohn N, Lesser M, Daum F. A multicenter trial of 6-
mercaptopurine and prednisone in children with newly diagnosed Crohn's disease.
Gastroenterology. 2000;119(4):895-902.
295. Coenen MJ, de Jong DJ, van Marrewijk CJ, Derijks LJ, Vermeulen SH, Wong DR, et al.
Identification of Patients With Variants in TPMT and Dose Reduction Reduces Hematologic
Events During Thiopurine Treatment of Inflammatory Bowel Disease. Gastroenterology.
2015;149(4):907-17.e7.
296. Newman WG, Payne K, Tricker K, Roberts SA, Fargher E, Pushpakom S, et al. A
pragmatic randomized controlled trial of thiopurine methyltransferase genotyping prior to
azathioprine treatment: the TARGET study. Pharmacogenomics. 2011;12(6):815-26.
297. Sayani FA, Prosser C, Bailey RJ, Jacobs P, Fedorak RN. Thiopurine methyltransferase
enzyme activity determination before treatment of inflammatory bowel disease with
azathioprine: effect on cost and adverse events. Can J Gastroenterol. 2005;19(3):147-51.
298. Patel V, Wang Y, MacDonald JK, McDonald JW, Chande N. Methotrexate for
maintenance of remission in Crohn's disease. Cochrane Database Syst Rev.
2014;2014(8):Cd006884.
299. Scherkenbach LA, Stumpf JL. Methotrexate for the Management of Crohn's Disease
in Children. Ann Pharmacother. 2016;50(1):60-9.
300. Grover Z, Burgess C, Muir R, Reilly C, Lewindon PJ. Early Mucosal Healing with
Exclusive Enteral Nutrition is Associated with Improved Outcomes in Newly Diagnosed
Children with Luminal Crohn's disease. J Crohns Colitis. 2016;10(10):1159-64.
301. Rutgeerts P, Van Assche G, Sandborn WJ, Wolf DC, Geboes K, Colombel JF, et al.
Adalimumab induces and maintains mucosal healing in patients with Crohn's disease: data
from the EXTEND trial. Gastroenterology. 2012;142(5):1102-11.e2.
302. Rutgeerts P, Diamond RH, Bala M, Olson A, Lichtenstein GR, Bao W, et al. Scheduled
maintenance treatment with infliximab is superior to episodic treatment for the healing of
mucosal ulceration associated with Crohn's disease. Gastrointest Endosc. 2006;63(3):433-
42; quiz 64.

95
303. Colombel JF, Sandborn WJ, Reinisch W, Mantzaris GJ, Kornbluth A, Rachmilewitz D,
et al. Infliximab, azathioprine, or combination therapy for Crohn's disease. N Engl J Med.
2010;362(15):1383-95.
304. Hazlewood GS, Rezaie A, Borman M, Panaccione R, Ghosh S, Seow CH, et al.
Comparative effectiveness of immunosuppressants and biologics for inducing and
maintaining remission in Crohn's disease: a network meta-analysis. Gastroenterology.
2015;148(2):344-54.e5; quiz e14-5.
305. Fan R, Zhong J, Wang ZT, Li SY, Zhou J, Tang YH. Evaluation of "top-down" treatment
of early Crohn's disease by double balloon enteroscopy. World J Gastroenterol.
2014;20(39):14479-87.
306. Matsumoto T, Motoya S, Watanabe K, Hisamatsu T, Nakase H, Yoshimura N, et al.
Adalimumab Monotherapy and a Combination with Azathioprine for Crohn's Disease: A
Prospective, Randomized Trial. J Crohns Colitis. 2016;10(11):1259-66.
307. Jones JL, Kaplan GG, Peyrin-Biroulet L, Baidoo L, Devlin S, Melmed GY, et al. Effects of
Concomitant Immunomodulator Therapy on Efficacy and Safety of Anti-Tumor Necrosis
Factor Therapy for Crohn's Disease: A Meta-analysis of Placebo-controlled Trials. Clin
Gastroenterol Hepatol. 2015;13(13):2233-40.e1-2; quiz e177-8.
308. Feagan BG, McDonald JW, Panaccione R, Enns RA, Bernstein CN, Ponich TP, et al.
Methotrexate in combination with infliximab is no more effective than infliximab alone in
patients with Crohn's disease. Gastroenterology. 2014;146(3):681-8.e1.
309. Kierkuś J, Iwańczak B, Wegner A, Dadalski M, Grzybowska-Chlebowczyk U, Łazowska
I, et al. Monotherapy with infliximab versus combination therapy in the maintenance of
clinical remission in children with moderate to severe Crohn disease. J Pediatr Gastroenterol
Nutr. 2015;60(5):580-5.
310. Moore C, Corbett G, Moss AC. Systematic Review and Meta-Analysis: Serum
Infliximab Levels During Maintenance Therapy and Outcomes in Inflammatory Bowel
Disease. J Crohns Colitis. 2016;10(5):619-25.
311. Nanda KS, Cheifetz AS, Moss AC. Impact of antibodies to infliximab on clinical
outcomes and serum infliximab levels in patients with inflammatory bowel disease (IBD): a
meta-analysis. Am J Gastroenterol. 2013;108(1):40-7; quiz 8.
312. Paul S, Moreau AC, Del Tedesco E, Rinaudo M, Phelip JM, Genin C, et al.
Pharmacokinetics of adalimumab in inflammatory bowel diseases: a systematic review and
meta-analysis. Inflamm Bowel Dis. 2014;20(7):1288-95.
313. Merras-Salmio L, Kolho KL. Clinical Use of Infliximab Trough Levels and Antibodies to
Infliximab in Pediatric Patients With Inflammatory Bowel Disease. J Pediatr Gastroenterol
Nutr. 2017;64(2):272-8.
314. Sandborn WJ, Gasink C, Gao LL, Blank MA, Johanns J, Guzzo C, et al. Ustekinumab
induction and maintenance therapy in refractory Crohn's disease. N Engl J Med.
2012;367(16):1519-28.

96

You might also like