Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2017-315866 on 18 May 2018. Downloaded from http://gut.bmj.com/ on August 30, 2021 at University of Maastricht Consortia. Protected by copyright.
Evolving role of diet in the pathogenesis and
treatment of inflammatory bowel diseases
Arie Levine,1,2 Rotem Sigall Boneh,1,2 Eytan Wine3

1
Pediatric Gastroenterology and Abstract
Nutrition Unit, Edith Wolfson Recent advances in basic and clinical science over the Significance of this study
Medical Center, Holon, Israel
2
Tel Aviv University, Tel Aviv, last 3 years have dramatically altered our appreciation
Israel of the role of diet in inflammatory bowel diseases What is already known?
3
Department of Pediatrics, (IBD). The marked increase in incidence of these ►► Diet has been implicated in the pathogenesis
University of Alberta, Edmonton, diseases along with the important role of non-genetic of Crohn disease (CD) and Ulcerative Colitis
Alberta, Canada (UC). Dietary factors may have a plausible role
susceptibility among patients with IBD has highlighted
that these diseases have a strong environmental in altering the microbiota, metabolome, host
Correspondence to barrier function and innate immunity.
Professor Arie Levine, Paediatric component. Progress in the field of microbiome and
Gastroenterology and Nutrition IBD has demonstrated that microbiome appears to What is new?
Unit, Wolfson Medical Center, play an important role in pathogenesis, and that diet ►► Exclusive enteral nutrition has demonstrated
Holon 58100 Israel; ​arie.​levine.​ may in turn impact the composition and functionality
dr@g​ mail.​com efficacy for induction of remission, as well
of the microbiome. Uncontrolled clinical studies have as treatment of complications such as
Received 2 January 2018 demonstrated that various dietary therapies such as inflammatory strictures, intrabdominal abscess
Revised 16 April 2018 exclusive enteral nutrition and newly developed exclusion and enterocutaneous fistulae in Crohn disease.
Accepted 17 April 2018 diets might be potent tools for induction of remission at Use or preoperative exclusive enteral nutrition
Published Online First disease onset, for patients failing biologic therapy, as a
18 May 2018 for ≥4 weeks may reduce the need for surgical
treatment for disease complications and in reducing the resections.
need for surgery. We review these advances from bench ►► Preliminary reports from uncontrolled studies
to bedside, along with the need for better clinical trials to suggest that specific exclusion diets based on
support these interventions. partial enteral nutrition and specific whole
foods may induce remission and can be used
as a salvage therapy after loss of response to
biologics or combination therapy.
Introduction
The basic understanding of how and why IBD How might it impact on clinical practice?
develop has been the focus of research of many ►► Dietary interventions ranging from dietary
disciplines over the last few decades, especially education of patients to exclusive enteral
given the complex, multifactorial nature of these nutrition and newer elimination diets may have
conditions. The discovery of NOD2 as the first, the potential to better control disease or avoid
and most important, susceptibility gene1 2 was then complications, pending better clinical trials.
followed by identification of over 200 other loci,3 4 There are insufficient data at the present time
but these genetic contributions together likely only to suggest that dietary therapy may be useful in
explain 19%–26% of the hereditary variance of UC; this is an important research gap.
IBD.5 Genetic studies have identified defects in
innate immunity in Crohn disease (CD) and altered
barrier function in Ulcerative Colitis (UC) as some hosts, in which bacteria for the most part do not
of the key players in disease susceptibility.6 7 reside adjacent to the mucosal epithelium (figure 1A,
The intestinal barrier comprises the mucous B).13 In fact, increased epithelial cell shedding, high
layer, epithelial cells and the tight junctions between vascular flow, depletion of the small bowel mucus
cells, rendering the mucosa relatively imperme- layer and presence of mucosa-associated bacteria
able to bacteria. Some of the early changes in the have been demonstrated in non-inflamed duodenum
sequence of events, leading to IBD are seen in the and ileum of children with UC.17–19 Both CD and UC
epithelial and mucus layers,8–10 which is particu- appear to involve translocation of bacteria through
larly evident in patients with UC.11 12 The mucous the epithelium.20 Thus, impaired barrier function,
layer is a critical barrier separating commensal and defects in innate immunity, changes in microbial
pathogenic bacteria from the epithelium, thereby composition and the presence of mucosa-associated
decreasing immune activation. Both CD and UC and translocating bacteria have all been described
are characterised by high penetration of the mucous in IBD. However, differences do exist between CD
layer by bacteria and increased barrier permea- (figure 1A) and UC (figure 1B).
bility.11 13 Increased barrier dysfunction during Dysbiosis is more prominent in CD.21 The
remission has been associated with clinical relapse predominance of Proteobacteria, specifically indi-
To cite: Levine A, Sigall of the disease.14 15 vidual Escherichia coli strains, especially in CD, has
Boneh R, Wine E. Gut CD and UC are characterised by the presence of been known for many years22 23; however, whether
2018;67:1726–1738. mucosal bacteria16 that differs from healthy human or not this reflects a true role in driving disease

1726   Levine A, et al. Gut 2018;67:1726–1738. doi:10.1136/gutjnl-2017-315866


Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2017-315866 on 18 May 2018. Downloaded from http://gut.bmj.com/ on August 30, 2021 at University of Maastricht Consortia. Protected by copyright.
Figure 1  Western diet and pathogenesis of IBD. (A) Proposed pathogenesis of Crohn disease and effects of western diet. (B) Proposed pathogenesis
of UC and effects of western diet observations that can be associated with western diet appear in the marked boxes. Tregs, regulatory T cells.

or just a change in the gut setting (eg, due to higher oxygen factors regulating compositional changes are access to calorie,
tension, related to inflammation) remains controversial; it is safe carbohydrate and protein intake.31 Since diet has such an important
to say that both processes are likely and that the diversity of role in gut homeostasis, and interacts with the microbiome, host
disease presentation and course likely represent different levels barrier and immunity, we will explore the possible and plausible
of microbial involvement in different individuals.24 The pres- role of dietary factors in the pathogenesis of IBD.
ence or decline in specific mucosal genera have been identified
in children and adults with IBD.21 25 26 Several researchers have Clues from epidemiology
demonstrated that patients with UC appear to have diminished Numerous epidemiological studies with inconsistent methodolog-
ability to produce short chain fatty acids (SCFA), even with an ical quality have demonstrated associations between the intake
abundant supply of substrates,27 28 suggesting that microbial of specific dietary components or dietary patterns and the risk of
metabolic dysfunction may play an important role in UC. developing IBD.32–39 One of the earliest epidemiological associ-
ations with IBD is the strong protective role of breast feeding.
Background: diet and IBD Although recall bias needs to be considered, almost all studies from
Diet is one of the key players in the normal gut microenvironment, various regions support this dose-dependent association, which is
impacting microbial composition, function, the gut barrier and also demonstrated for other immune-mediated conditions.40 The
host immunity. Alterations in single food groups have far ranging results from other epidemiological studies should be considered
implications. For example, depriving colonic microbes from fibre with caution due to a recall bias and possible dietary modification
in rodent models will favour taxa that can use alternative carbon before the diagnosis of IBD.41 A statistical association between
sources found within the colonic mucus; this then leads to depletion specific dietary factors and disease does not necessarily imply
of the mucus layer, disruption of the barrier, immune activation and causation. In any complex disease with dietary triggers, it may be
tissue damage.29 Changing from a plant-based to an animal-based difficult to unravel the role of individual dietary risk factors since
diet radically changes bacterial taxa and metabolism, leading to dietary patterns often involve exposures to clusters of foods. In
alterations in bile acids and sulfide metabolism, both of which may addition, certain dietary ingredients are likely to cluster together
play a role in IBD.30 Recent data suggest that the most important due to industrial processing of food. For example, fat exposure
Levine A, et al. Gut 2018;67:1726–1738. doi:10.1136/gutjnl-2017-315866 1727
Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2017-315866 on 18 May 2018. Downloaded from http://gut.bmj.com/ on August 30, 2021 at University of Maastricht Consortia. Protected by copyright.
will likely be associated with emulsifier exposure, processed meats of the intact mucous layer, decreased regulatory T cells (Tregs) and
with preservatives, dairy products with thickening agents and impaired bacterial clearance mechanisms (figure 1A, B). A major
emulsifiers, etc. In addition, food frequency questionnaires may limitation to date is that most of the evidence comes from animal
be insufficient to address longitudinal exposures. models and human cell lines, which makes extrapolation difficult.
D’Souza et al conducted a case-control study to assess the dietary Dietary factors that may affect host immunity are presented in
patterns of Canadian children and the risk of developing CD. They table 1 and figure 2. Western diet is characterised by alterations
evaluated multiple dietary patterns including western diet and a in the amount of exposure to natural dietary ingredients such as
prudent diet. A dietary pattern characterised by meats, fatty foods high fat (HF), high sugar (HS), high wheat and high dairy expo-
and desserts (western diet) increased the risk for CD in females (OR sure, accompanied by low fibre exposure, and by high exposure
4.7, 95% CI 1.6 to 14.2), whereas the consumption of vegetables, to multiple food additives.45 46 We will focus our discussion on
fruits, olive oil, fish, grains and nuts (prudent diet) was associated the dietary products with the most evidence from animal models,
with a decreased risk for developing CD in both males and females which include fibre, animal fat, wheat and food additives.
(females: OR 0.3, 95% CI 0.1 to 0.9; males: OR 0.2, 95% CI 0.1 Low dietary fibre, identified through epidemiological studies
to 0.5).33 A recent Australian population-based study of environ- as a possible risk factor,33 37 could affect host immunity via
mental risk factors demonstrated an increased risk for CD with multiple pathways. Fibres and starches found in fruits and vege-
frequent consumption of fast food (western diet) before diagnosis.42 tables are a vital substrate for the production of butyrate and
Several studies have associated protein with increased risk for other SCFAs. Butyrate plays an important role in downregulating
IBD.32 35 36 As part of the E3N prospective study, which included inflammation by suppressing transcription of cytokines and
67 581 French women, high animal protein was associated with increasing differentiation and the population of lamina propria
a significantly increased risk of IBD (OR 3.03, 95% CI 1.45 to Tregs.47 48 Butyrate can also enhance innate immunity by upreg-
6.34), particularly with UC (OR 3.29, 95% CI 1.34 to 8.04).32 ulating defensins and cathelicidins in animal models.47 48 Low
Vegetable protein intake was not associated with either disease. dietary fibre may cause catabolism of the mucous layer, leading
However, other smaller studies have not confirmed this.43 In a to increased permeability of the mucous layer, and allowing
separate study evaluating dietary intake and relapse of UC, red increased contact between luminal bacteria and the epithelium.29
meat was found to have the strongest association (OR 5.19, HF or HF/HS diets may act synergistically to a low fibre diet
95% CI 2.09 to 12.9) with relapse.36 as they may exert many of the same effects described above for
Data regarding the associations between increased intakes of low fibre exposure. An HF/HS diet in a CEACAM 10 rodent
carbohydrates and the risk for IBD are inconsistent. The E3N model has been shown to decrease MUC2 expression and goblet
study did not identify carbohydrate intake as a risk factor for cell mucous while increasing intestinal permeability.49 These
IBD, CD or UC.32 Racine et al, evaluated 366 351 participants diets have been demonstrated to decrease both butyrate produc-
with IBD from theEuropean Prospective Investigation into tion and expression of the butyrate GPR43 receptor, which is
Cancer and Nutrition (EPIC) study cohort and found no correla- underexpressed in CD.49 Finally, HF diets (without HS) have
tion between dietary intake patterns and CD.34 They did find been shown to increase tumour necrosis factor (TNF)-α and
an association between high-sugar and low-fibre intake and UC interferon-γ expression and decrease levels of colonic Tregs.50
(OR 1.68, 95% CI 1.00 to 2.82). Exposure to gluten may promote an inflammatory state, impair
More recent data from the EPIC study found no associa- innate immunity or increase intestinal permeability. Dietary
tion between alcohol consumption and the risk for CD.44 This gluten significantly decreased intestinal Tregs in non-obese
contrasted with the study by Jowett et al,36 who tried to find diabetic mice fed gluten compared with standard chow.51 Dietary
dietary products associated with the relapse of UC. High alcohol gluten was also shown to induce ileitis in TNFΔARE/WT mice
consumption was associated with an increased risk of relapse via an increase in intestinal permeability,52 which has been iden-
(OR 2.71, 95% CI 1.1 to 6.67). tified as a factor associated with relapse in CD.14
Dietary fats are associated with an increase or decrease risk Alpha-amylase/trypsin inhibitors, found in wheat, may have a
for IBD, depending on the type of fat. Among 170 805 women pro-inflammatory role. They are activators of human dendritic cells
followed over 26 years, a high intake of dietary long-chain n-3 and macrophages, driving intestinal inflammation in rodents.53 54
polyunsaturated fatty acids was associated with a reduced risk of Multiple food additives, commonly present in western diet,
UC. In contrast, high intake of transunsaturated fats was associ- may affect host barrier function or immunity. Chassaing et al,
ated with an increased risk of UC.37 demonstrated that IL-10−/− mice exposed to two common emul-
Perhaps the dietary component with the greatest agreement in sifiers, carboxymethylcellulose and polysorbate-80, developed
epidemiological studies is dietary fibre, which is also represented increased intestinal permeability and significant thinning of the
in the prudent dietary patterns mentioned above. Multiple mucous layer, leading to increased proximity of the microbiota
studies reported a negative association between dietary fibre to the intestinal epithelium.55 Maltodextrins, used as thickeners
intake from fruits or fruits and vegetables and subsequent risk and sweeteners, have demonstrated the ability to affect mucosal
for CD.33 37 Data from the Nurses’ Health Study among 170 776 proximity (by a presumed effect on mucous layer thickness)
participants showed that higher long-term intake of fruit was and to impair clearance of intracellular Salmonella.56 57 Carra-
associated with lower risk of CD but not UC.37 geenans, used for texture and as thickening agents in the dairy
Several research gaps remain in this field, including more data and sauce industry, may induce intestinal permeability.58 59 Other
about exposure to industry-added ingredients, such as emulsi- factors, which may promote intestinal permeability are listed in
fiers, preservatives and thickening agents, including maltodex- table 1 and are beyond the scope of this paper.
trins and carrageenans.
Diet and microbiota
Dietary components linked to changes in the microbiome that
Diet and altered host immunity have been associated with IBD appear in table 2 and figure 3. For
Host factors that may play an important role in pathogenesis or example, adherent invasive E. coli (AIEC) strains cannot induce
active inflammation include defects in barrier function, depletion inflammation under normal conditions. Pathogenicity may arise
1728 Levine A, et al. Gut 2018;67:1726–1738. doi:10.1136/gutjnl-2017-315866
Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2017-315866 on 18 May 2018. Downloaded from http://gut.bmj.com/ on August 30, 2021 at University of Maastricht Consortia. Protected by copyright.
Table 1  Dietary factors potentially affecting host barrier and immunity
Dietary component Reference Model Effect
Natural components
 High fat+high sugar diet Martinez-Medina. Gut, CEABAC 10 mice ►► Decreased MUC2 expression
201449 ►► Depleted Goblet cells
►► Increased intestinal permeability
►► Increased TNF-α secretion
 High fat+high sugar diet Agus. Sci Rep, 201660 CEABAC 10 mice ►► Decreased FoxP3 Tregs in mesenteric lymph nodes
►► Decreased butyrate production
►► Reducedexpression of the butyrate GPR43 receptor
►► Dysbiosis
►► Significantly decreased SCFA concentrations
 High fat diet Ma. Clin Exp Immunol, WT C57BL/6 mice ►► Higher numbers of non-CD1d-restricted natural killer T cells in the colonic
200850 IEL
►► Increased TNF-α and IFN-γ expression
►► Decreased levels of colonic Tregs
 High fat diet Suzuki. Nutri & Metab, LETO and ►► Increased intestinal permeability
2010101 OLETF rats (mutant obese Long-Evans) ►► Decreased tight junction proteins expression (claudin-1, claudin-3, occludin
Human intestinal Caco-2 cells and junctional adhesion molecule-1)
 High fat diet Gruber. PLoS One, 2013102 TNFΔARE/WT mouse and WT C57BL/6 ►► Aggravation of ileal inflammation
►► Reduced expression of occludin
►► Increasedtranslocation of endotoxin
►► Increased pro-inflammatory markers
►► Recruitment of dendritic cells and Th17-biased lymphocyte into the lamina
propria
 Sodium caprate Söderholm. Dig Dis Sci, Rat ileum ►► Increased tight junction permeability
(constituent of milk fat) 1998103
Söderholm. Gut, 2002104 Specimens from the distal ileum of ►► Rapid increased in paracellular permeability
patients with CD
 Gluten Ejsing-Duun. Scand J NOD and BALB/c mice ►► Decreased the occurrence of Tregs by 10%–15% (p<0.05)
Immunol, 200851
 Gluten Wagner. Inflamm Bowel TNFΔARE/WT mouse ►► Increased intestinal permeability
Dis, 201352 ►► Reduced occludin expression
►► Induced chronic ileitis
 Gluten (gliadin) Lammers. Human intestinal epithelial cell, Caco2 ►► Zonulin released, causedtight junction disassembly
Gastroenterology, 2008105 cells
IEC6 cells
 Gluten (gliadin) Hollon. Nutrients, 2015106 Ex vivo human duodenal biopsies ►► Increased intestinal permeability
►► Higher concentration of IL-10 in controls compared with coeliac disease in
remission or gluten sensitivity
 Wheat (ATIs) Junkers. J Exp Med, Human cells and biopsies ►► ATIs are potent activators of human innate immune responses in
201254 monocytes, macrophages and dendritic cells
 Wheat (ATIs) Zevallos. TLR4-responsive mouse and human ►► Activation of dendritic cells in mesenteric lymph nodes
Gastroenterology, 201753 cell lines ►► Released inflammatory mediators
 Soluble fibres and resistant Bassaganya-Riera. J Nutr, C57BL/6J WT mice ►► Decreased ileal and colonic inflammatory lesions
starch 2011107 IL-10−/− mice ►► Decreased IFN-γ production by effector CD4+ T cells from Peyer’s patches
►► Resistant starch increased the IL-10-expressing cells
►► Suppressed gut inflammation
 Dietary pectin Ye. J Agric Food Chem, IL-10−/− mice ►► Reduced expression of TNF-R and GATA-3
2010108 ►► Lower levels of IgE, IgG and IgM expression
►► Modulation of production of pro-inflammatory cytokines and
immunoglobulins
 Fermentable fibre (guar Hung. J Nutr, 2016109 BALB/c mice aged 7 weeks ►► Increased expression of occludin and claudin-3, claudin-4 and claudin-7
gum, partially hydrolysed (reduced permeability)
GG) ►► Greater total faecal SCFA concentrations
►► Reduced inflammation score
 Multifibre mix diet Wang. JCC, 201647 IL-10−/− mice ►► Reduced disease activity index score
►► Decreased CD4+CD45+ lymphocytes, IFN-γ/IL-17A, TNF-α/TNF-R2 mRNA
expression
►► Increased Tregs and SCFA production
►► Increased epithelial expression and correct localisation of tight junction
proteins
 Alcohol Forsyth. Alcohol Clin Exp Mice ►► Colonic (but not small intestinal) hyperpermeability
Res, 2017110 ►► Decreased butyrate/total SCFA ratio in stool
 Dietary salt Tubbs. J Immunol, 2017111 IL-10−/− murine model of colitis ►► Exacerbation of inflammatory pathology
►► Enhanced expression of numerous pro-inflammatory cytokines
►► TLR4 activation

Continued
Levine A, et al. Gut 2018;67:1726–1738. doi:10.1136/gutjnl-2017-315866 1729
Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2017-315866 on 18 May 2018. Downloaded from http://gut.bmj.com/ on August 30, 2021 at University of Maastricht Consortia. Protected by copyright.
Table 1  Continued 
Dietary component Reference Model Effect
Food additives
 Carboxymethylcellulose Chassaing. Nature, IL-10−/− mice ►► Increased intestinal permeability
(E466) and polysorbate-80 201555 ►► Mucous layer depletion led to increased proximity of the microbiota to the
(E433) intestinal epithelium
 Carboxymethylcellulose Swidsinski. Inflamm IL-10−/− mice ►► Distension of spaces between villi, with bacteria filling these spaces
(E466) Bowel Dis, 2009112
 Maltodextrin Miyazato. Biosci BALB/c mice ►► Increasedtotal IgA levels in the intestinal tract
Microbiota Food Health,
2016113
 Carrageenan (E407) Choi. Toxicol Lett, 201259 HCT-8, HT-29 and Caco-2 cells ►► Lowered transepithelial resistance
►► Discontinuous and irregular ZO-1 expression
Fahoum. Mol Nutr Food Caco-2 cell model ►► Redistribution of the tight-junction protein ZO-1
Res, 201758 ►► Increased intestinal permeability
 Titanium dioxide (TiO2) Ruiz. Gut, 2017114 WT and NLRP3−/− mice with DSS ►► Oral administration of TiO2 worsened acute colitis through a mechanism
involving the NLRP3 inflammasome
►► Induced reactive oxygen species generation
►► Increased epithelial permeability in IEC monolayers
ATI, α-a mylase/trypsin inhibitors; CD, Crohn disease; IFN, interferon; IL, interleukin; SCFA, short chain fatty acids; Th, T helper; TNF, tumour necrosis factor; Tregs, T regulatory cells;
WT, wild-type; ZO, zonula occludens. IEL (intrepithelial lymphocyes), DSS (dextran sulfate sodium).

if the ligand CEACAM 6 is presented by intestinal epithelial and by an increase in Proteobacteria and a decrease in Firmicutes,
M cells. Under these conditions, AIEC may form small intestinal similar to that seen in CD.61 HF diets result in accumulation of
biofilms, adhere and translocate via M cells, and then invade the secondary bile acids, such as deoxycholic acid, which in turn can
associated epithelium. An HF/HS diet may promote colonisation inhibit the growth of members of the Bacteroidetes and Firmic-
with IBD-associated pathobionts. CEABAC 10 mice, expressing utes phyla, characteristic of the dysbiosis found in CD.62
CEACAM 6, fed HF/HS but not standard chow were rapidly Maltodextrins have been shown to promote AIEC biofilm
colonised by mucosa-associated AIEC and presented higher formation independent of the presence of the ligand CEACAM 6,
degrees of crypt abscesses when compared with the standard while AIEC growth in medium was enhanced particularly with
chow group.49 Transplantation of faeces from HF/HS-treated polysaccharides used as thickening agents, such as maltodextrin
mice to germ free mice increased susceptibility to AIEC.60 A and xanthan gum, but not by glucose or sucrose.63
study that compared HF with HS diet in a mouse model demon- The source of dietary fat may play an important role in
strated that HF but not HS induced dysbiosis, characterised promoting colitogenic bacteria. Devkota et al reported an

Figure 2  Possible effects of dietary factors on host barrier and immunity leading to IBD (based on cell lines and animal models). Tregs, regulatory T cells.
1730 Levine A, et al. Gut 2018;67:1726–1738. doi:10.1136/gutjnl-2017-315866
Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2017-315866 on 18 May 2018. Downloaded from http://gut.bmj.com/ on August 30, 2021 at University of Maastricht Consortia. Protected by copyright.
Table 2  Dietary factors potentially affecting the microbiota in IBD
Dietary component Reference Model Effect
Natural components
 High fat+high sugar diet Martinez-Medina. Gut, 201449 CEABAC 10 mice ►► AIEC Colonisation
►► Low diversity
►► Dysbiosis
►► Higher degree of crypt abscesses
 High fat+high sugar diet Agus. Sci Rep, 201660 CEABAC 10 mice ►► Intestinal mucosa dysbiosis
►► Decreased butyrate production
►► Reduced expression of the butyrate GPR43 receptor
►► Overgrowth of Escherichia coli
►► Significantly decreased SCFA concentrations
 High fat vs high sugar Lai. Environ Pollut, 201661 CD1 mice ►► High fat but not high sugar induced dysbiosis characterised by an increase
in Proteobacteria and a decrease in Firmicutes and Clostridia
►► Growth induction of the family Helicobacteraceae
 Milk fat (saturated fat) Devkota. Nature, 201264 IL-10−/− mice ►► Promoted expansion of sulfite-reducing pathobiont, Bilophila
wadsworthia
►► Induced pro-inflammatory Th1 immune response
►► Increased incidence of colitis
►► Low diversity
►► Dysbiosis
 High fibre diet Silveira. Eur J Nutr, 2017115 BALB/c female mice ►► High fibre diet protected from acute colitis
 Fibre James. GUT, 201528 Patients with UC and ►► High fibre diet did not increased SCFA production in patients with UC
controls
 Animal vs plant diet David. Nature, 201430 Healthy volunteers ►► Animal-based diet increased the abundance of bile-
tolerant  microorganisms and decreased the levels of Firmicutes
 Alcohol Forsyth. Alcohol Clin Exp Res, Mice ►► Decreased butyrate/total SCFA ratio in stool
2017110
Food additives
 Carboxymethylcellulose Chassaing. Nature, 201555 IL-10–/– mice ►► Increased mucosal-associated bacteria
(E466) and polysorbate-80 Chassaing. GUT, 201766 Human intestinal microbial ►► Increased Flagellin expression
(E433) ecosystem (M-SHIME) ►► P-80 altered species composition
 Carboxymethylcellulose Swidsinski. Inflamm Bowel Dis, IL-10–/– mice ►► Bacterial overgrowth
(E466) 2009112 ►► Distension of spaces between villi, with bacteria filling these spaces
►► Adherence of bacteria to the mucosa
►► Migration of bacteria to the bottom of the crypts of Lieberkühn
 Polysorbate-80 Roberts. Gut, 201065 M cells ►► Increasedtranslocation of E. coli across M cells
 (E433) Caco-2 cells
 Plantain and broccoli fibre Human Peyer’s patches ►► Reduction in translocation of E. coli across M cells
 Maltodextrin Nickerson. PLoS One, 201263 Human intestinal epithelial ►► Biofilm formation AIEC
Nickerson. Gut Microbes, 201556 cell monolayers ►► Enhanced E. coli adhesion independent of the cellular receptor CEACAM 6
 Carrageenan (E407) Munyaka. Front Microbiol, Piglet model of IBD ►► Decreased bacterial species richness
2016116 ►► Shifted community composition.
►► Bacterial dysbiosis
AIEC, adherent invasive E. coli ; SCFA, short chain fatty acids; Th, T helper.

increase in colitis severity after exposure to milk fat diet but not The metabolic activity of the microbiota may be as important,
to isocaloric polyunsaturated fatty acid diet or a low fat diet in or even more important than compositional changes, especially in
IL-10−/− mice.64 Colitis was associated with the bloom of coli- UC. Both CD and UC are characterised by a decrease in SCFA-pro-
togenic Bilophila wadsworthia in the milk fat group, which was ducing genera, which are stimulated by the presence of dietary fibre.
dependent on milk fat, and induced taurine conjugated bile acids. However, UC seems to be characterised by deficient production of
Interestingly, emulsifiers may affect the microbiome as well as the SCFA, even when substrates such as resistant starch are provided.27 28
host. The emulsifier polysorbate-80 was associated with increased Sulfide-reducing bacteria and increased sulfide production have been
translocation of AIEC through M cells and the follicular associated reported in UC.27 Animal protein-based diets favour production of
epithelium; this effect was suppressed by certain dietary fibres.65 sulfide reductases and sulfide-reducing bacteria.30
Emulsifiers appear to affect compositional changes associated with In summary, high animal or dairy fat, animal protein, wheat,
inflammation. Polysorbate-80 induced inflammation and flagellin emulsifiers and thickeners appear to top the list of candidate
expression via alteration of microbial dysbiosis, whereas the emul- foods associated with intestinal inflammation in animal models.
sifier carboxymethylcellulose rapidly induced flagellin and alter-
ations in gene expression for flagellin in a model for gut dysbiosis.66 Diet as a therapeutic intervention
Exclusion of western diet by exclusive enteral nutrition (EEN) or an Recent studies have expanded our understanding of the possible
exclusion diet in children with CD were shown to cause a marked roles for dietary therapy, primarily in CD. Dietary therapies, such
reduction in Proteobacteria and in taxa associated previously with as EEN, were traditionally and primarily used to induce remis-
CD including Escherichia, Haemophilus, Veillonella and Fusobac- sion in early or new-onset CD67 or as supportive therapy in chil-
teria  (Van Limbergen and Dunn, unpublished data). dren and adults with malnutrition or in the preoperative setting.
Levine A, et al. Gut 2018;67:1726–1738. doi:10.1136/gutjnl-2017-315866 1731
Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2017-315866 on 18 May 2018. Downloaded from http://gut.bmj.com/ on August 30, 2021 at University of Maastricht Consortia. Protected by copyright.
Figure 3  Possible effects of dietary factors on microbiota leading to IBD (based on cell lines and animal models).

EEN involves the exclusive use of a liquid nutrition in the form Use of EEN poses challenges to physicians and patients
of medical formulas, without exposure to other foods, usually for alike, as patients have to deal with monotony of food and
6–8 weeks (table 3). This effect does not depend on the protein taste fatigue. Furthermore, up to 50% of patients may require
source (type of formula), but is very dependent on exclusion of nasogastric tubes and many might refuse to start therapy
ordinary table food.68 69 Newer open-label studies have demon- due to these issues.74 75 This limits access and availability of
strated that existing and new dietary strategies may have a wider the therapy. Therefore, it is no surprise that the last 3 years
role and can be employed to maintain remission, induce remission have seen several attempts to achieve the effect of EEN while
in patients failing biologics and for patients with complicated disease allowing access to whole foods. Investigators from three
(figure 4). As data from randomised controlled trials (RCTs) are centres in the USA and Canada compared outcomes from three
absent or pending, we will confine ourselves to more recent studies cohorts treated with either: 1) high volume partial enteral
that have expanded our understanding beyond the use of EEN at nutrition (PEN) using a mean of 77% of estimated energy
diagnosis for induction of remission, which is reviewed elsewhere.67 requirements from PEN with free access to food; 2) EEN and
3) infliximab. Clinical remission rates were similar between
Induction of remission infliximab and EEN (73%–76%) and lower (50%) in the PEN
Several recent paediatric studies have demonstrated and confirmed group. Faecal calprotectin <250 g/dL as an inflammatory end
that EEN may induce remission in 60%–86% of children accom- point was found only in 14% of patients treated with PEN
panied by a significant decrease in inflammatory markers such as and ad libitum diet, compared with 45% with EEN and 62%
erythrocyte sedimentation rate (ESR), C reactive protein (CRP) treated with infliximab at week 8. This suggests that high-
and faecal calprotectin.69–73 This recent body of literature has volume PEN with access to free diet is ineffective at improving
evaluated long-term benefits associated with use of EEN in mild- inflammation compared with EEN or infliximab, reaffirming
to-moderate paediatric CD. In comparison to use of steroids and the principle of exclusivity.67 68
corrected for disease severity, EEN was associated with higher Other research groups have employed a different strategy based
remission rates, better growth and longer steroid-free periods,71 72 on the combination of PEN with a specific exclusion diet that
but did not differ with regard to other outcomes such as relapse. allows access to whole foods. This review will be confined only to
Cohen-Dolev et al demonstrated that EEN was superior to studies that evaluated clinical remission and objective markers of
corticosteroids for induction of remission and linear growth in inflammation or mucosa healing. The Crohn’s disease exclusion
a multicentre prospective inception cohort of mild-to-moderate diet was developed based on the principles outlined in figures 1–3,
CD at diagnosis. EEN was not superior with respect to other by excluding foods that have been associated with altered host
outcomes such as time to relapse or complications over 2 years.72 barrier or bacterial clearance, dysbiosis and virulence factors
Grover et al performed a prospective trial to assess mucosal that may allow bacteria to become mucosa-associated and enable
healing by performing colonoscopy before and after 8 weeks of translocation, based on the bacterial penetration cycle hypothesis
EEN with azathioprine. At the end of 8 weeks, complete mucosal for CD first proposed by the authors.76 The premise behind this
healing was observed in 33%, near complete in another 19% of theory is that CD is caused by mucosal and translocating bacteria.
patients.73 Patients who achieved complete mucosal healing had This diet was evaluated initially in 47 children and adults with
better sustained remission over 3 years of follow-up. mild-to-moderate disease. Clinical remission was achieved in

1732 Levine A, et al. Gut 2018;67:1726–1738. doi:10.1136/gutjnl-2017-315866


Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2017-315866 on 18 May 2018. Downloaded from http://gut.bmj.com/ on August 30, 2021 at University of Maastricht Consortia. Protected by copyright.
Table 3  Key studies and recent advances with EEN in children and adults
Duration of
Study design Population EEN Comparator Number of patients Outcome Key findings Reference
Randomised Adults 3–6 Weeks EEN vs CS and EEN: 51 pts Improvement in CDAI: Equal effectiveness of Malchow. Scand
controlled trial sulfasalazine Drugs: 44 pts EEN: 41% EEN compared with J Gastroenterol,
Drugs: 72% drugs per protocol 1990117
p<0.05
Randomised Adults 4–6 Weeks EEN vs CS and EEN: 52 pts Remission: Comparison to Lochs.
ccontrolled trial sulfasalazine Drugs: 55 pts EEN: 55% medications Gastroenterology,
Drugs: 74% 1991118
p<0.01
Randomised Adults 4 Weeks EEN: n-6 PUFA vs MUFA: 20 pts Remission rates (ITT): Composition of fat Gassull. Gut,
ccontrolled trial MUFA vs CS with PUFA: 23 pts MUFA EEN: 20% 2002119
ward diet CS: 19 pts PUFA EEN: 52%
CS: 79%
p=0.001
Randomised Paediatric CD 6 Weeks Elemental formula vs Elemental fomula: 16 pts Remission: Polymeric equivalent to Ludvigsson. Acta
ccontrolled trial polymeric formula Polymeric formula: 17 pts Elemental: 69% elemental Paediatr, 2004120
Polymeric: 82%
p=0.44
Randomised Paediatric CD 6 Weeks EEN vs 50% PEN EEN:24 pts Remission: Principle of exclusivity Johnson. GUT,
ccontrolled trial with free diet PEN: 26 pts PEN: 15% 200668
EEN: 42%
p=0.035
Randomised Newly diagnosed 10 Weeks Polymeric formula EEN: 19 pts Remission: Mucosal healing Borrelli. Clin
ccontrolled trial paediatric CD vs CS CS: 18 pts polymeric EEN: 79% comparing EEN with CS Gastroenterol
CS: 67% Hepatol, 2006121
p=0.4
Mucosal healing EEN: 74%
CS: 33%
p<0.05
Retrospective Paediatric CD 8 Weeks Oral vs continues Oral: 45 pts Remission: Oral equivalent to Rubio. Aliment
enteral feeding Enteral: 61 pts Oral EEN: 75% enteral feeding Pharmacol Ther,
Tube EEN: 85% 2011122
p=0.157
Prospective Adults 4 Weeks – EEN: 13 pts Significant improvement in Improvement of quality Gou. Nutr Clin
uncontrolled quality of life of life Pract, 2013123
Clinical remission: 84.6%
Prospective Newly diagnosed 6–8 Weeks EEN vs CS mild-to- 5-ASA: 29 pts CS-free remission week 12: EEN superior to CS Levine. IBD, 201470
inception cohort children moderate CD EEN: 43 pts EEN: 71% for reduction of
CS: 114 pts CS: 46% inflammation
5-ASA: 55%
p=0.0006
NCR:
EEN: 39%
CS: 25%
5-ASA: 24%
Prospective cohort Paediatric CD 6 Weeks EEN vs healthy EEN: 15 pts EEN: decreased butyrate, EEN mechanism Gerasimidis. IBD,
control HC: 21 pts decreased diversity, 2014124
Faecalibacterium prausnitzii
concentration decreased
Prospective cohort Newly diagnosed 6 Weeks Mild-to-moderate 34 Children Clinical remission: 84% Mucosal healing Grover. J
paediatric CD EEN with thiopurine Early good endoscopic Gastroenterol,
response: 58% 2014125
Complete transmural healing
ileal CD: 21%
Prospective cohort Paediatric CD 8 Weeks EEN vs anti-TNF vs Anti-TNF: 52 pts Clinical response: PEN- 64% Comparison with anti- Lee. IBD, 201569
PEN with ad libitum EEN: 22 pts EEN- 88% TNF
diet PEN: 16 pts Anti-TNF - 84% EEN reduced
p=0.08 FCP<250 mg/g: calprotectin
PEN: 14%
EEN -45%
Anti-TNF- 62%
p=0.001
Retrospective Newly diagnosed 8–16 Weeks EEN vs CS EEN:76 pts EEN: 86.6% remission vs CS: Reduced need for Connors. JCC,
paediatric CD CS 35 pts 58.1% steroids 201771
p<0.01 
Prospective Newly diagnosed 6–8 Weeks EEN vs CS EEN: 60 pts Remission: EEN superior to CS Cohen Dolev. JCC,
inception cohort children CS: 87 pts CS: 47% 201872
EEN: 63%
p=0.036 
5-ASA, 5-aminosalicylic acid; CD, Crohn disease; CDAI, Crohn’s Disease Activity Index; CS, corticosteroids; EEN, exclusion enteral nutrition; FCP, faecal calprotectin; ITT, intention to treat;
MUFA, monounsaturated fatty acids; NCR, normal C reactive protein remission; PEN, partial enteral nutrition; PUFA, polyunsaturated fatty acids.

Levine A, et al. Gut 2018;67:1726–1738. doi:10.1136/gutjnl-2017-315866 1733


Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2017-315866 on 18 May 2018. Downloaded from http://gut.bmj.com/ on August 30, 2021 at University of Maastricht Consortia. Protected by copyright.
Figure 4  New targets for dietary interventions in Crohn disease (CD).

70% of patients; this was accompanied by a significant drop in is established, it is certainly possible that diet will be sufficient to
ESR and CRP and was associated with mucosal healing among maintain homeostasis and prevent the cascade of pro-inflamma-
patients who achieved remission and were evaluated during the tory events leading to flare; however, long-term studies are chal-
maintenance phase of the diet.77 The diet is low in animal fat, lenging to perform and adherence becomes a serious concern.
rich in complex carbohydrates, with moderate exposure to soluble Nevertheless, several studies have assessed PEN for maintenance
fibre. It excludes wheat and dairy, emulsifiers, maltodextrins, cara- of remission, some showing promising results.
geenans and sulfites. This diet was subsequently evaluated for One of the first studies to suggest the merit of PEN was
induction of remission in children and adults failing biologics. published in Gut in 1996. Wilschanski et al82 followed 28 children
Among 21 patients with active disease after loss of response to who responded to EEN induction therapy for up to 12 months and
biologics despite dose escalation or combination therapy, 61% agreed to receive nightly nasogastric PEN; they were less likely to
achieved clinical remission, again with a significant drop in inflam- flare than the 19 patients who chose not to receive PEN. A review,
matory markers.78 While limited by design, these two retrospec- which included 10 published studies, most of which were not
tive studies reported remission with an exclusion diet at both ends randomised, demonstrated some endoscopic improvement with
of the spectrum of clinical disease, suggesting that diet may be an PEN, but the overall quality of these studies was limited, high-
important tool for controlling active disease even in established lighting the need for large, prospective RCTs.83
disease and among biologic unresponsive patients. These reports A retrospective, single-centre study from Israel followed 42
should be viewed as preliminary data that need to be validated children who responded to EEN and then went on PEN (50%
in prospective controlled trials. Several RCTs using this method liquid formula, 50% free diet) for a median of 40 months.
are ongoing at the present time (NCT01728870, NCT02843100, Although a survival analysis was not included, median time for
NCT02231814). The specific carbohydrate diet restricting carbo- remaining in remission was 6 months; however, the median time
hydrates and processed foods has been advocated for CD and in remission without need for additional therapy was 0 months
UC. This was previously evaluated in 10 children with very mild (0–16 months). Eighty-six percent of patients started on PEN as
CD (mean Harvey Bradshaw Index (HBI) 3.3, mean ESR 9.7), maintenance therapy required additional therapy by 6 months.84
of whom 6 achieved clinical remission.79 However, another small There is insufficient evidence to recommend PEN as the only
study published this year demonstrated that this diet was not asso- maintenance therapy in children based on existing data.
ciated with mucosal healing.80 Clinical trials to assess this diet are Another approach, mainly based on studies in adults from
underway at the present time (NCT02213835, NCT01749813), Japan, has focused on partial elemental diet (ED), providing a
but randomised controlled studies evaluating patients with more minimum of 900 kcal/day combined with a free diet. An RCT
significant disease are required. Fibre and prebiotics supplements comparing this approach to a fully free diet showed lower relapse
have been evaluated as interventions in CD. A systematic review rates at a mean follow-up of 1 year (34.6% vs 64%).85 More
including 10 eligible studies with clinical outcomes found no recent retrospective studies have indicated a potential advan-
evidence for use of fibre or prebiotics to induce or maintain remis- tage for partial ED in reducing loss of response to infliximab
sion in CD.81 Importantly, these papers concluded that there was and adalimumab.86 87 PEN has also been shown to be effective in
no evidence for restriction of fibre in the absence of obstructive preventing postoperative recurrence of CD. Twenty adults who
symptoms or stricturing disease. were compliant to EN were treated with nightly ED and a low fat
diet during the day and were followed for 5 years; remarkably, 16
Maintenance of remission maintained the diet. Only two had disease recurrence (vs 9/20 in
The idea of maintaining remission with diet is compelling and a normal-diet control group; p=0.03) and 1 (vs 5) required addi-
challenging at the same time. On the one hand, once remission tional surgery.88 Japanese researchers randomised adult patients

1734 Levine A, et al. Gut 2018;67:1726–1738. doi:10.1136/gutjnl-2017-315866


Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2017-315866 on 18 May 2018. Downloaded from http://gut.bmj.com/ on August 30, 2021 at University of Maastricht Consortia. Protected by copyright.
with CD in remission on maintenance 5-aminosalicylic acid carbohydrates (low fermentable oligosaccharides, disaccharides,
(ASA) to additional 6-mercaptopurine (MP), PEN with 900 kcal/ monosaccharides and polyols (FODMAP) diet) is effective in the
day from an ED, or no additional therapy. At the end of 2 years, management of functional symptoms. Prince et al found that
sustained remission was present in 60% of patients on 6-MP, 47% 78%, p<0.001 with IBD reported satisfactory relief of func-
of patients on PEN and only 27% of patients on 5-ASA. Both tional symptoms after 6 weeks of low FODMAP diet.98 A recent
6-MP and PEN were significantly superior to 5-ASA.89 Fernán- meta-analysis concluded that a low FODMAP diet is beneficial
dez-Bañares et al demonstrated that combining fibre supplements for reducing GI symptoms in patients with quiescent IBD.99
with mesalamine was more effective than mesalamine alone for Halmos et al100 showed that the low FODMAP diet had no effect
maintenance of remission.90 A systematic review found that there on the calprotectin results in patients with CD. Furthermore,
was no evidence for an effect of dietary fibre on disease outcomes altering dietary FODMAP intake was associated with possible
in UC.81 detrimental changes in faecal microbiota and therefore is not
recommended for extended periods of time.

Dietary therapy for complicated CD


CD is associated with several complications that eventually can Summary
lead to surgery. Treatment of complicated disease, such as stric- In summary, there seems to be a growing body of evidence to
tures, abscesses and fistulae has traditionally been either medical suggest that dietary factors may play an important role in the patho-
or surgical. Recent studies have demonstrated that dietary genesis of CD. Use of EEN has expanded from a tool for induction
therapy with EEN may have the potential to treat complications of remission in children at disease onset, to a tool with potential
and have a ‘surgery sparing effect’. to treat very complicated long duration disease in adults (figure 4).
Two studies conducted by a group from China evaluated the Use of perioperative EEN has the potential to avoid the need for
efficacy of EEN in complicated CD. Hu et al conducted a prospec- surgery, reduce poor surgical outcomes and delay postoperative
tive, observational study to examine the effects of 12 weeks of recurrence in CD. Perhaps the most exciting development is the
EEN on inflammatory bowel strictures. They performed cross-sec- potential to induce and maintain remission by use of exclusion diets
tional CT before and after therapy. Sixty-five per cent of patients and PEN, which would enable wider use of dietary therapy across
achieved clinical remission, while 54% achieved radiologic remis- the spectrum. However, the field still has insufficient high-quality
sion. This was accompanied by a significant increase in luminal evidence due to the small number of well-designed trials performed
diameter and a significant decrease in bowel wall thickness.91 Yan to date, but the future appears to be bright in CD. RCTs with newer
et al conducted a prospective study to identify the predictors of dietary interventions or indications are warranted. Perhaps the
response to 12 weeks of EEN in patients with CD who had entero- largest research gaps at the present are in the field of UC. Although
cutaneous fistulae. They demonstrated that 30/48 (62.5%) had there are emerging clues linking diet to UC as well, researchers have
a successfully closure of fistula after EEN therapy with average yet to make the jump from bench to bedside in UC, and we still do
closure time of 32.4±8.85 days. They found that decreased CRP not know if dietary therapy would be effective in UC.
and elevated body mass index levels were associated with response Contributors  All authors contributed to the writing of the manuscript.
to EN in patients with CD with enterocutaneous fistulae.92
Funding  This study was funded by grants from Nestle and the Azrieli Foundation.
EEN has also been demonstrated to reduce the need for surgery
in complicated disease. Heerasing et al demonstrated in a retro- Competing interests  None declared.
spective case-control study that EEN for a period of 6 weeks could Patient consent  Not required.
lead to avoidance of surgery in (13/51) 25% of patients who were Provenance and peer review  Commissioned; externally peer reviewed.
scheduled for an elective surgical resection.93 They also showed by © Article author(s) (or their employer(s) unless otherwise stated in the text of the
multivariable logistic regression analysis that patients who went to article) 2018. All rights reserved. No commercial use is permitted unless otherwise
surgery without EEN had a ninefold increase in the incidence of expressly granted.
postoperative abscess and/or anastomotic leak (OR 9.1; 95% CI
1.2 to 71.2, p=0.04). An additional recent retrospective study
conducted by a group from China reported that patients with CD References
with intra-abdominal abscesses who were treated with 4 weeks of 1 Ogura Y, Bonen DK, Inohara N, et al. A frameshift mutation in NOD2 associated with
susceptibility to Crohn’s disease. Nature 2001;411:603–6.
EEN and antibiotics were less likely to require surgical interven-
2 Hugot J-P, Chamaillard M, Zouali H, et al. Association of NOD2 leucine-rich repeat
tion compared with those who did not use EEN (26.1% vs 56.3%, variants with susceptibility to Crohn’s disease. Nature 2001;411:599–603.
p=0.01).94 Several studies have confirmed that perioperative EEN 3 Luo Y, de Lange KM, Jostins L, et al. Exploring the genetic architecture of
can improve surgical outcomes, reduce complications and the need inflammatory bowel disease by whole-genome sequencing identifies association at
for immune suppression.95 96 Use of preoperative EEN may have ADCY7. Nat Genet 2017;49:186–92.
long-lasting effects. Wang et al found that EEN prior to resection 4 Huang H, Fang M, Jostins L, et al. Fine-mapping inflammatory bowel disease loci to
single-variant resolution. Nature 2017;547:173–8.
was accompanied by a significant reduction of endoscopic recur- 5 Peters LA, Perrigoue J, Mortha A, et al. A functional genomics predictive
rence rates after resection for CD after 6 months.96 In addition, a network model identifies regulators of inflammatory bowel disease. Nat Genet
large retrospective study demonstrated that administration of EEN 2017;49:1437–49.
for approximately 4 weeks led to lower rates of stoma creation 6 Jostins L, Ripke S, Weersma RK, et al. Host–microbe interactions have shaped the
(p<0.05), a decrease in urgent operation requirement (p<0.05) genetic architecture of inflammatory bowel disease. Nature 2012;491:119–24.
7 Bording-Jorgensen M, Alipour M, Danesh G, et al. Inflammasome Activation by ATP
and extended preoperative drug-free intervals (p<0.001).97
Enhances Citrobacter rodentium Clearance through ROS Generation. Cell Physiol
Biochem 2017;41:193–204.
8 Teshima CW, Goodman KJ, El-Kalla M, et al. Increased intestinal permeability
Diet for functional symptoms
in relatives of patients with crohn’s disease is not associated with small bowel
Patients with IBD may experience GI symptoms even during ulcerations. Clinical Gastroenterology and Hepatology 2017;15:1413–8.
remission without underlying inflammation. Recent studies have 9 Buhner S, et al. Genetic basis for increased intestinal permeability in families with
demonstrated that restriction of several groups of fermentable Crohn’s disease: role of CARD15 3020insC mutation? Gut 2006;55:342–7.

Levine A, et al. Gut 2018;67:1726–1738. doi:10.1136/gutjnl-2017-315866 1735


Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2017-315866 on 18 May 2018. Downloaded from http://gut.bmj.com/ on August 30, 2021 at University of Maastricht Consortia. Protected by copyright.
10 Kevans D, Turpin W, Madsen K, et al. Determinants of intestinal permeability in 40 Xu L, Lochhead P, Ko Y, et al. Systematic review with meta-analysis: breastfeeding
healthy first-degree relatives of individuals with crohnʼs disease. Inflamm Bowel Dis and the risk of Crohn’s disease and ulcerative colitis. Aliment Pharmacol Ther
2015;21:879–87. 2017;46:780–9.
11 Johansson MEV, Gustafsson JK, Holmén-Larsson J, et al. Bacteria penetrate the 41 Loftus EV. Clinical epidemiology of inflammatory bowel disease: incidence,
normally impenetrable inner colon mucus layer in both murine colitis models and prevalence, and environmental influences. Gastroenterology 2004;126:1504–17.
patients with ulcerative colitis. Gut 2014;63:281–91. 42 Niewiadomski O, Studd C, Wilson J, et al. Influence of food and lifestyle on the risk
12 McCole DF. IBD candidate genes and intestinal barrier regulation. Inflamm Bowel Dis of developing inflammatory bowel disease. Intern Med J 2016;46:669–76.
2014;20:1829–49. 43 Hou JK, Abraham B, El-Serag H. Dietary intake and risk of developing inflammatory
13 Kleessen B, Kroesen AJ, Buhr HJ, et al. Mucosal and invading bacteria in patients bowel disease: a systematic review of the literature. Am J Gastroenterol
with inflammatory bowel disease compared with controls. Scand J Gastroenterol 2011;106:563–73.
2002;37:1034–41. 44 Bergmann MM, Hernandez V, Bernigau W, et al. No association of alcohol use and
14 Kiesslich R, Duckworth CA, Moussata D, et al. Local barrier dysfunction identified by the risk of ulcerative colitis or Crohn’s disease: data from a European Prospective
confocal laser endomicroscopy predicts relapse in inflammatory bowel disease. Gut cohort study (EPIC). Eur J Clin Nutr 2017;71:512–8.
2012;61:1146–53. 45 Moubarac J-C, Batal M, Martins APB, et al. Processed and ultra-processed food
15 Vogelsang H. Do changes in intestinal permeability predict disease relapse in crohnʼs products: consumption trends in Canada from 1938 to 2011. Canadian Journal of
Dietetic Practice and Research 2014;75:15–21.
disease? Inflamm Bowel Dis 2008;14(Suppl 2):S162–3.
46 Roberts CL, Rushworth SL, Richman E, et al. Hypothesis: Increased consumption of
16 Gevers D, Kugathasan S, Knights D, et al. A microbiome foundation for the study of
emulsifiers as an explanation for the rising incidence of Crohn’s disease. Journal of
crohn’s disease. Cell Host Microbe 2017;21:301–4.
Crohn's and Colitis 2013;7:338–41.
17 Zaidi D, Bording-Jorgensen M, Huynh HQ, et al. Increased epithelial gap density in
47 Wang H, Shi P, Zuo L, et al. Dietary non-digestible polysaccharides ameliorate
the noninflamed duodenum of children with inflammatory bowel diseases. J Pediatr
intestinal epithelial barrier dysfunction in IL-10 Knockout Mice. Journal of Crohn's
Gastroenterol Nutr 2016;63:644–50.
and Colitis 2016;10:1076–86.
18 Zaidi D, Churchill L, Huynh HQ, et al. Capillary flow rates in the duodenum of
48 Furusawa Y, Obata Y, Fukuda S, et al. Commensal microbe-derived butyrate induces
pediatric ulcerative colitis patients are increased and unrelated to inflammation. J the differentiation of colonic regulatory T cells. Nature 2013;504:446–50.
Pediatr Gastroenterol Nutr 2017;65:306–10. 49 Martinez-Medina M, Denizot J, Dreux N, et al. Western diet induces dysbiosis with
19 Alipour M, Zaidi D, Valcheva R, et al. Mucosal barrier depletion and loss of bacterial increased E coli in CEABAC10 mice, alters host barrier function favouring AIEC
diversity are primary abnormalities in paediatric ulcerative colitis. Journal of Crohn's colonisation. Gut 2014;63:116–24.
and Colitis 2016;10:462–71. 50 Ma X, Torbenson M, Hamad ARA, et al. High-fat diet modulates non-CD1d-
20 Vrakas S, Mountzouris KC, Michalopoulos G, et al. Intestinal bacteria composition restricted natural killer T cells and regulatory T cells in mouse colon and exacerbates
and translocation of bacteria in inflammatory bowel disease. PLoS One experimental colitis. Clinical & Experimental Immunology 2008;151:130–8.
2017;12:e0170034. 51 Ejsing-Duun M, Josephsen J, Aasted B, et al. Dietary gluten reduces the number of
21 Pascal V, Pozuelo M, Borruel N, et al. A microbial signature for Crohn’s disease. Gut intestinal regulatory t cells in mice. Scand J Immunol 2008;67:553–9.
2017;66:813–22. 52 Wagner SJ, Schmidt A, Effenberger MJP, et al. Semisynthetic diet ameliorates crohn’s
22 Mazzarella G, Perna A, Marano A, et al. Pathogenic role of associated adherent- Disease–Like Ileitis in TNFΔARE/WT mice through antigen-independent mechanisms
invasive Escherichia coli in Crohn’s Disease. J Cell Physiol 2017;232:2860–8. of gluten. Inflamm Bowel Dis 2013;19:1285–94.
23 Darfeuille-Michaud A, Boudeau J, Bulois P, et al. High prevalence of adherent- 53 Zevallos VF, Raker V, Tenzer S, et al. Nutritional Wheat Amylase-Trypsin Inhibitors
invasive Escherichia coli associated with ileal mucosa in Crohn’s disease. Promote Intestinal Inflammation via Activation of Myeloid Cells. Gastroenterology
Gastroenterology 2004;127:412–21. 2017;152:1100–13.
24 Sartor RB, Wu GD, Gd W. Roles for intestinal bacteria, viruses, and fungi in 54 Junker Y, Zeissig S, Kim S-J, et al. Wheat amylase trypsin inhibitors drive intestinal
pathogenesis of inflammatory bowel diseases and therapeutic approaches. inflammation via activation of toll-like receptor 4. J Exp Med 2012;209:2395–408.
Gastroenterology 2017;152:327–39. 55 Chassaing B, Koren O, Goodrich JK, et al. Dietary emulsifiers impact the mouse gut
25 Gevers D, Kugathasan S, Denson LA, et al. The treatment-naive microbiome in new- microbiota promoting colitis and metabolic syndrome. Nature 2015;519:92–6.
onset crohn’s disease. Cell Host Microbe 2014;15:382–92. 56 Nickerson KP, Chanin R, McDonald C. Deregulation of intestinal anti-microbial
26 Machiels K, Sabino J, Vandermosten L, et al. Specific members of the predominant gut defense by the dietary additive, maltodextrin. Gut Microbes 2015;6:78–83.
microbiota predict pouchitis following colectomy and IPAA in UC. Gut 2017;66:79–88. 57 Nickerson KP, Homer CR, Kessler SP, et al. The dietary polysaccharide maltodextrin
27 Khalil NA, Walton GE, Gibson GR, et al. In vitro batch cultures of gut microbiota from promotes salmonella survival and mucosal colonization in mice. PLoS One
healthy and ulcerative colitis (UC) subjects suggest that sulphate-reducing bacteria 2014;9:e101789.
levels are raised in UC and by a protein-rich diet. Int J Food Sci Nutr 2014;65:79–88. 58 Fahoum L, Moscovici A, David S, et al. Digestive fate of dietary carrageenan:
28 James SL, Christophersen CT, Bird AR, et al. Abnormal fibre usage in UC in remission. evidence of interference with digestive proteolysis and disruption of gut epithelial
Gut 2015;64:562–70. function. Mol Nutr Food Res 2017;61:16.
29 Desai MS, Seekatz AM, Koropatkin NM, et al. A dietary fiber-deprived gut microbiota 59 Choi HJ, Kim J, Park S-H, et al. Pro-inflammatory NF-κB and early growth response
degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell gene 1 regulate epithelial barrier disruption by food additive carrageenan in human
2016;167:1339–53. intestinal epithelial cells. Toxicol Lett 2012;211:289–95.
30 David LA, Maurice CF, Carmody RN, et al. Diet rapidly and reproducibly alters the 60 Agus A, Denizot J, Thévenot J, et al. Western diet induces a shift in microbiota
human gut microbiome. Nature 2014;505:559–63. composition enhancing susceptibility to Adherent-Invasive E. coli infection and
31 Holmes AJ, Chew YV, Colakoglu F, et al. Diet-Microbiome interactions in health are intestinal inflammation. Sci Rep 2016;6:19032.
controlled by intestinal nitrogen source constraints. Cell Metab 2017;25:140–51. 61 Lai K-P, Chung Y-T, Li R, et al. Bisphenol A alters gut microbiome: Comparative
32 Jantchou P, Morois S, Clavel-Chapelon F, et al. Animal protein intake and risk metagenomics analysis. Environ Pollut 2016;218:923–30.
of inflammatory bowel disease: the E3N prospective study. Am J Gastroenterol 62 Islam KBMS, Fukiya S, Hagio M, et al. Bile acid is a host factor that regulates the
2010;105:2195–201. composition of the cecal microbiota in rats. Gastroenterology 2011;141:1773–81.
33 DʼSouza S, Levy E, Mack D, et al. Dietary patterns and risk for Crohnʼs disease in 63 Nickerson KP, McDonald C. Crohn’s disease-associated adherent-invasive escherichia
children. Inflamm Bowel Dis 2008;14:367–73. coli adhesion is enhanced by exposure to the Ubiquitous Dietary Polysaccharide
34 Racine A, Carbonnel F, Chan SS, et al. Dietary Patterns and risk of inflammatory Maltodextrin. PLoS One 2012;7:e52132.
bowel disease in Europe: results from the EPIC Study. Inflamm Bowel Dis 64 Devkota S, Wang Y, Musch MW, et al. Dietary-fat-induced taurocholic
2016;22:345–54. acid promotes pathobiont expansion and colitis in Il10 −/− mice. Nature
35 Shoda R, Matsueda K, Yamato S, et al. Epidemiologic analysis of Crohn disease in Japan: 2012;487:104–8.
increased dietary intake of n-6 polyunsaturated fatty acids and animal protein relates to 65 Roberts CL, Keita AV, Duncan SH, et al. Translocation of Crohn’s disease Escherichia
the increased incidence of Crohn disease in Japan. Am J Clin Nutr 1996;63:741–5. coli across M-cells: contrasting effects of soluble plant fibres and emulsifiers. Gut
36 Jowett SL, et al. Influence of dietary factors on the clinical course of ulcerative colitis: 2010;59:1331–9.
a prospective cohort study. Gut 2004;53:1479–84. 66 Chassaing B, Van de Wiele T, De Bodt J, et al. Dietary emulsifiers directly alter
37 Ananthakrishnan AN, Khalili H, Konijeti GG, et al. A prospective study of long- human microbiota composition and gene expression ex vivo potentiating intestinal
term intake of dietary fiber and risk of crohn’s disease and ulcerative colitis. inflammation. Gut 2017;66:1414–27.
Gastroenterology 2013;145:970–7. 67 Ruemmele FM, Veres G, Kolho KL, et al. Consensus guidelines of ECCO/ESPGHAN on
38 Costea I, Mack DR, Lemaitre RN, et al. Interactions between the dietary the medical management of pediatric Crohn’s disease. Journal of Crohn's and Colitis
polyunsaturated fatty acid ratio and genetic factors determine susceptibility to 2014;8:1179–207.
pediatric crohn’s disease. Gastroenterology 2014;146:929–31. 68 Johnson T, Macdonald S, Hill SM, et al. Treatment of active Crohn’s disease in
39 Chan SSM, Luben R, van Schaik F, et al. Carbohydrate intake in the etiology of children using partial enteral nutrition with liquid formula: a randomised controlled
crohnʼs disease and ulcerative colitis. Inflamm Bowel Dis 2014;20:2013–21. trial. Gut 2006;55:356–61.

1736 Levine A, et al. Gut 2018;67:1726–1738. doi:10.1136/gutjnl-2017-315866


Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2017-315866 on 18 May 2018. Downloaded from http://gut.bmj.com/ on August 30, 2021 at University of Maastricht Consortia. Protected by copyright.
69 Lee D, Baldassano RN, Otley AR, et al. Comparative effectiveness of nutritional and 94 Zheng XB, Peng X, Xie XY, et al. Enteral nutrition is associated with a decreased
biological therapy in north american children with active crohnʼs disease. Inflamm risk of surgical intervention in Crohn’s disease patients with spontaneous intra-
Bowel Dis 2015;21:1786–93. abdominal abscess. Rev Esp Enferm Dig 2017;109:109.
70 Levine A, Turner D, Pfeffer Gik T, et al. Comparison of outcomes parameters for 95 Li G, Ren J, Wang G, et al. Preoperative exclusive enteral nutrition reduces the
induction of remission in new onset pediatric Crohn’s disease: evaluation of the postoperative septic complications of fistulizing Crohn’s disease. Eur J Clin Nutr
porto IBD group "growth relapse and outcomes with therapy" (GROWTH CD) study. 2014;68:441–6.
Inflamm Bowel Dis 2014;20:278–85. 96 Wang H, Zuo L, Zhao J, et al. Impact of Preoperative Exclusive Enteral Nutrition on
71 Connors J, Basseri S, Grant A, et al. Exclusive enteral nutrition therapy in paediatric Postoperative Complications and Recurrence After Bowel Resection in Patients with
crohn’s disease results in long-term avoidance of corticosteroids: results of Active Crohn’s Disease. World J Surg 2016;40:1993–2000.
a propensity-score matched cohort analysis. Journal of Crohn's and Colitis 97 Li Y, Zuo L, Zhu W, et al. Role of exclusive enteral nutrition in the preoperative
2017;11:1063–70. optimization of patients with Crohn’s disease following immunosuppressive therapy.
72 Cohen-Dolev N, Sladek M, Hussey S, et al. Differences in outcomes over time with Medicine 2015;94:e478.
exclusive enteral nutrition compared with steroids in children with mild to moderate 98 Prince AC, Myers CE, Joyce T, et al. Fermentable Carbohydrate Restriction (Low
crohn’s disease: results from the GROWTH CD Study. Journal of Crohn's and Colitis FODMAP Diet) in clinical practice improves functional gastrointestinal symptoms in
2018;12:306–12. patients with inflammatory bowel disease. Inflamm Bowel Dis 2016;22:1129–36.
73 Grover Z, Burgess C, Muir R, et al. Early mucosal healing with exclusive enteral 99 Zhan YL, Zhan YA, Dai SX. Is a low FODMAP diet beneficial for patients with
nutrition is associated with improved outcomes in newly diagnosed children with inflammatory bowel disease? A meta-analysis and systematic review. Clin Nutr
luminal crohn’s disease. Journal of Crohn's and Colitis 2016;10:1159–64. 2018;37:123–9.
74 Buchanan E, Gaunt WW, Cardigan T, et al. The use of exclusive enteral nutrition for 100 Halmos EP, Christophersen CT, Bird AR, et al. Consistent prebiotic effect on
induction of remission in children with Crohn’s disease demonstrates that disease gut microbiota with altered FODMAP intake in patients with crohn’s disease: a
phenotype does not influence clinical remission. Aliment Pharmacol Ther 2009;30:501–7. randomised, controlled cross-over trial of well-defined diets. Clin Transl Gastroenterol
75 Van Limbergen J, Haskett J, Griffiths AM, et al. Toward enteral nutrition in 2016;7:e164.
the treatment of pediatric crohn disease in canada: a workshop to identify 101 Suzuki T, Hara H. Dietary fat and bile juice, but not obesity, are responsible
barriers and enablers. Canadian Journal of Gastroenterology and Hepatology for the increase in small intestinal permeability induced through the suppression
2015;29:351–6. of tight junction protein expression in LETO and OLETF rats. Nutr Metab
76 Levine A, Wine E. Effects of enteral nutrition on Crohn’s disease: clues to the impact 2010;7:19.
of diet on disease pathogenesis. Inflamm Bowel Dis 2013;19:1322–9. 102 Gruber L, Kisling S, Lichti P, et al. High fat diet accelerates pathogenesis of murine
77 Sigall-Boneh R, Pfeffer-Gik T, Segal I, et al. Partial enteral nutrition with a Crohn’s crohn’s disease-like ileitis independently of obesity. PLoS One 2013;8:e71661.
disease exclusion diet is effective for induction of remission in children and young 103 Soderholm JD, Oman H, Blomquist L, et al. Reversible increase in tight junction
adults with Crohn’s disease. Inflamm Bowel Dis 2014;20:1353–60. permeability to macromolecules in rat ileal mucosa in vitro by sodium caprate, a
78 Sigall Boneh R, Sarbagili Shabat C, Yanai H, et al. Dietary therapy with the crohn’s constituent of milk fat. Dig Dis Sci 1998;43:1547–52.
disease exclusion diet is a successful strategy for induction of remission in children 104 Soderholm JD, et al. Augmented increase in tight junction permeability by luminal
and adults failing biological therapy. J Crohns Colitis 2017;11:1205–12. stimuli in the non-inflamed ileum of Crohn’s disease. Gut 2002;50:307–13.
79 Cohen SA, Gold BD, Oliva S, et al. Clinical and mucosal improvement with 105 Lammers KM, Lu R, Brownley J, et al. Gliadin induces an increase in intestinal
specific carbohydrate diet in pediatric Crohn disease. J Pediatr Gastroenterol Nutr permeability and zonulin release by binding to the chemokine receptor CXCR3.
2014;59:516–21. Gastroenterology 2008;135:194–204.
80 Wahbeh GT, Ward BT, Lee DY, et al. Lack of Mucosal Healing From Modified Specific 106 Hollon J, Puppa E, Greenwald B, et al. Effect of gliadin on permeability of intestinal
Carbohydrate Diet in Pediatric Patients With Crohn Disease. J Pediatr Gastroenterol biopsy explants from celiac disease patients and patients with non-celiac gluten
Nutr 2017;65:289–92. sensitivity. Nutrients 2015;7:1565–76.
81 Wedlake L, Slack N, Andreyev HJ, et al. Fiber in the treatment and maintenance of 107 Bassaganya-Riera J, DiGuardo M, Viladomiu M, et al. Soluble fibers and resistant
inflammatory bowel disease: a systematic review of randomized controlled trials. starch ameliorate disease activity in interleukin-10–deficient mice with inflammatory
Inflamm Bowel Dis 2014;20:576–86. bowel disease. J Nutr 2011;141:1318–25.
82 Wilschanski M, Sherman P, Pencharz P, et al. Supplementary enteral nutrition 108 Mb Y, Lim BO. Dietary pectin regulates the levels of inflammatory cytokines and
maintains remission in paediatric Crohn’s disease. Gut 1996;38:543–8. immunoglobulins in interleukin-10 knockout mice. Journal of agricultural and food
83 Yamamoto T, Nakahigashi M, Umegae S, et al. Enteral nutrition for the maintenance chemistry 2010;58:11281–6.
of remission in Crohn’s disease: a systematic review. Eur J Gastroenterol Hepatol 109 Hung TV, Suzuki T. Dietary Fermentable Fiber Reduces Intestinal Barrier Defects and
2010;22:1–8. Inflammation in Colitic Mice. J Nutr 2016;146:1970–9.
84 Schulman JM, Pritzker L, Shaoul R. Maintenance of remission with partial 110 Forsyth CB, Shaikh M, Bishehsari F, et al. Alcohol feeding in mice promotes colonic
enteral nutrition therapy in pediatric crohn’s disease: a retrospective study. Can J hyperpermeability and changes in colonic organoid stem cell fate. Alcoholism:
Gastroenterol Hepatol 2017;2017:1–7. Clinical and Experimental Research 2017;41:2100–13.
85 Takagi S, Utsunomiya K, Kuriyama S, et al. Effectiveness of an ’half elemental diet’ 111 Tubbs AL, Liu B, Rogers TD, et al. Dietary salt exacerbates experimental colitis. The
as maintenance therapy for Crohn’s disease: A randomized-controlled trial. Aliment Journal of Immunology 2017;199:1051–9.
Pharmacol Ther 2006;24:1333–40. 112 Swidsinski A, Ung V, Sydora BC, et al. Bacterial overgrowth and inflammation of
86 Sugita N, Watanabe K, Kamata N, et al. Efficacy of a concomitant elemental diet small intestine after carboxymethylcellulose ingestion in genetically susceptible mice.
to reduce the loss of response to adalimumab in patients with intractable Crohn’s Inflamm Bowel Dis 2009;15:359–64.
disease. J Gastroenterol Hepatol 2018;33:631-637. 113 Miyazato S, Kishimoto Y, Takahashi K, et al. Continuous intake of resistant
87 Kamata N, Oshitani N, Watanabe K, et al. Efficacy of concomitant elemental diet maltodextrin enhanced intestinal immune response through changes in the intestinal
therapy in scheduled infliximab therapy in patients with Crohn’s disease to prevent environment in mice. Biosci Microbiota Food Health 2016;35:1–7.
loss of response. Dig Dis Sci 2015;60:1382–8. 114 Ruiz PA, Morón B, Becker HM, et al. Titanium dioxide nanoparticles exacerbate DSS-
88 Yamamoto T, Shiraki M, Nakahigashi M, et al. Enteral nutrition to suppress induced colitis: role of the NLRP3 inflammasome. Gut 2017;66:1216–24.
postoperative Crohn’s disease recurrence: a five-year prospective cohort study. Int J 115 Silveira ALM, Ferreira AVM, de Oliveira MC, et al. Preventive rather than
Colorectal Dis 2013;28:335–40. therapeutic treatment with high fiber diet attenuates clinical and inflammatory
89 Hanai H, Iida T, Takeuchi K, et al. Nutritional therapy versus 6-mercaptopurine markers of acute and chronic DSS-induced colitis in mice. Eur J Nutr
as maintenance therapy in patients with Crohn’s disease. Dig Liver Dis 2017;56:179–91.
2012;44:649–54. 116 Munyaka PM, Sepehri S, Ghia J-E, et al. Carrageenan gum and adherent invasive
90 Fernández-Bañares F, Hinojosa J, Sánchez-Lombraña JL, et al. Randomized clinical Escherichia coli in a piglet model of inflammatory bowel disease: impact on
trial of Plantago ovata seeds (dietary fiber) as compared with mesalamine in intestinal mucosa-associated microbiota. Front Microbiol 2016;7:462.
maintaining remission in ulcerative colitis. Spanish Group for the Study of Crohn’s 117 Malchow H, Steinhardt HJ, Lorenz-Meyer H, et al. Feasibility and
Disease and Ulcerative Colitis (GETECCU). Am J Gastroenterol 1999;94:427–33. effectiveness of a defined-formula diet regimen in treating active Crohn’s
91 Hu D, Ren J, Wang G, et al. Exclusive enteral nutritional therapy can relieve disease. European Cooperative Crohn’s Disease Study III. Scand J Gastroenterol
inflammatory bowel stricture in Crohn’s disease. J Clin Gastroenterol 1990;25:235–44.
2014;48:790–5. 118 Lochs H, Steinhardt HJ, Klaus-Wentz B, et al. Comparison of enteral nutrition and
92 Yan D, Ren J, Wang G, et al. Predictors of response to enteral nutrition in drug treatment in active Crohn’s disease. Results of the European Cooperative
abdominal enterocutaneous fistula patients with Crohn’s disease. Eur J Clin Nutr Crohn’s Disease Study. IV. Gastroenterology 1991;101:881–8.
2014;68:959–63. 119 Gassull MA, Fernandez-Banares F, Cabre E, et al. Fat composition may be a
93 Heerasing N, Thompson B, Hendy P, et al. Exclusive enteral nutrition provides an clue to explain the primary therapeutic effect of enteral nutrition in Crohn’s
effective bridge to safer interval elective surgery for adults with Crohn’s disease. disease: results of a double blind randomised multicentre European trial. Gut
Aliment Pharmacol Ther 2017;45:660–9. 2002;51:164–8.

Levine A, et al. Gut 2018;67:1726–1738. doi:10.1136/gutjnl-2017-315866 1737


Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2017-315866 on 18 May 2018. Downloaded from http://gut.bmj.com/ on August 30, 2021 at University of Maastricht Consortia. Protected by copyright.
120 Ludvigsson JF, Krantz M, Bodin L, et al. Elemental versus polymeric enteral nutrition 123 Guo Z, Wu R, Zhu W, et al. Effect of Exclusive enteral nutrition on health-related
in paediatric Crohn’s disease: a multicentre randomized controlled trial. Acta quality of life for adults with active crohn’s disease. Nutrition in Clinical Practice
Paediatr 2004;93:327–35. 2013;28:499–505.
121 Borrelli O, Cordischi L, Cirulli M, et al. Polymeric diet alone versus 124 Gerasimidis K, Bertz M, Hanske L, et al. Decline in presumptively protective gut
corticosteroids in the treatment of active pediatric crohn’s disease: a randomized bacterial species and metabolites are paradoxically associated with disease
controlled open-label trial. Clinical Gastroenterology and Hepatology improvement in pediatric crohn’s disease during enteral nutrition. Inflamm Bowel Dis
2006;4:744–53. 2014;20:861–71.
122 Rubio A, Pigneur B, Garnier-Lengliné H, et al. The efficacy of exclusive nutritional 125 Grover Z, Muir R, Lewindon P. Exclusive enteral nutrition induces early clinical,
therapy in paediatric Crohn’s disease, comparing fractionated oral vs. continuous mucosal and transmural remission in paediatric Crohn’s disease. J Gastroenterol
enteral feeding. Aliment Pharmacol Ther 2011;33:1332–9. 2014;49:638–45.

1738 Levine A, et al. Gut 2018;67:1726–1738. doi:10.1136/gutjnl-2017-315866

You might also like