You are on page 1of 11

Journal of Crohn's and Colitis, 2022, 16, 852–862

https://doi.org/10.1093/ecco-jcc/jjab212
Advance access publication 22 November 2021
Review Article

Inflammatory Bowel Disease-associated Fatty Liver


Disease: the Potential Effect of Biologic Agents
Apostolis Papaefthymiou,a,b, Spyros Potamianos,a Antonis Goulas,b Michael Doulberis,b,c
Jannis Kountouras,d, Stergios A. Polyzosb,

Downloaded from https://academic.oup.com/ecco-jcc/article/16/5/852/6433262 by ECCO Member Access user on 24 June 2022


a
Department of Gastroenterology, University Hospital of Larisa, Larisa, Thessaly, Greece
b
First Laboratory of Pharmacology, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
c
Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, Aarau, Switzerland
d
Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
Corresponding author: Apostolis Papaefthymiou, MD, PhD Candidate, First Laboratory of Pharmacology, Aristotle University of Thessaloniki, Campus of
Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece. Tel.: +302310999316, +306973853042; email: apostokp@auth.gr

Abstract
Inflammatory bowel diseases [IBD] exhibit intestinal and systemic manifestations. Nonalcoholic fatty liver disease [NAFLD] is a common
co-existing condition, possibly contributing to the cardio-metabolic burden and overall morbidity. Εmerging therapeutic choices of biologic agents
have modified the clinical course of IBD; however, their impact on IBD-associated NAFLD has not been extensively evaluated. The prevalence
of NAFLD varies among IBD patients, but it appears higher than in the general population in the majority of quality studies. In terms of patho-
genetic and risk factors of NAFLD, they may vary with IBD activity. Dysbiosis, mucosal damage, and cytokine release have been implicated in
the pathogenesis during the relapses, whereas metabolic risk factors seem to play a dominant role during the remissions of IBD. Considering
biologics, although quality data are scarce, agents suppressing tumour necrosis factor may offer potential benefits in IBD-associated NAFLD,
whereas anti-integrins do not appear to confer any therapeutic advantage.
In conclusion, IBD-associated NAFLD possibly follows two different patterns, one manifested during the relapses and one during the remissions
of IBD. Some, but not all, biologics may benefit NAFLD in patients with IBD. Further mechanistic and prospective cohort studies are warranted
to illuminate the effects of various biologics on NAFLD.
Key Words: Biologics; inflammatory bowel diseases; nonalcoholic fatty liver disease

1. Introduction IBD-associated parameters, including disease activity and dur-


Inflammatory bowel diseases [IBD], i.e., Crohn’s disease [CD] and ation and prior surgical intervention, may also impact on the de-
ulcerative colitis [UC], represent multifactorial disorders of the velopment of NAFLD.10 Non-metabolic factors, such as chronic
gastrointestinal [GI] tract with a broad spectrum of clinical mani- uncontrolled inflammation, use of glucocorticosteroids, pro-
festations. The introduction of biological treatments [or biologics] longed starvation, rapid weight loss or uncontrolled refeeding,
has modified the natural history of CD and UC and expanded the and sarcopenia have also been proposed to contribute to the
periods of remission.1 Despite their GI origin, IBD have been as- pathogenesis of NAFLD in patients with IBD.11–14
sociated with a plethora of extra-intestinal manifestations.2 In this Despite its high prevalence, there is to date no approved
regard, nonalcoholic fatty liver disease [NAFLD] has been linked medication for NAFLD.15 On the other hand, the treatment
to IBD, representing a common diagnosis during the workup for choices for IBD have been expanded after the introduction of
elevated liver function tests [LFTs] among patients with IBD.3,4 biologics16 which seem to optimise the therapeutic schemes,
NAFLD has a global prevalence of 25% in the general thus improving the control of disease activity in the long term
population, being much higher in specific subgroups, e.g., pa- as well as its systemic manifestations. Nonetheless, the potential
tients with obesity and/or type 2 diabetes mellitus [T2DM].5 serious adverse effects of biologics on patients with IBD should
NAFLD refers to a phenotypic spectrum, starting from sim- be carefully weighed before and during their clinical use.17
ple steatosis or nonalcoholic fatty liver [NAFL] which may The above considering, this narrative review aims to sum-
progress to nonalcoholic steatohepatitis [NASH], hepatic marise evidence on the association between IBD and NAFLD,
fibrosis, cirrhosis, and hepatocellular carcinoma in a mi- as well as on the potential effects of biologics having been
nority of patients.6 Of note, the additive burden of NAFLD introduced in the treatment of IBD, on NAFLD.
in patients with IBD has been associated with a 2-fold risk
of cardiovascular events and a 5-fold risk of chronic kidney
2. Evidence Linking IBD with NAFLD
disease [CKD], thus increasing overall mortality.7,8 NAFLD is
a multifactorial disease that may affect, but also be affected 2.1. Prevalence of NAFLD in IBD
by, the components of metabolic syndrome [MetS] including A broad range of NAFLD prevalence has been reported in
obesity, T2DM, dyslipidaemia, and arterial hypertension.9 various studies, probably due to the different populations

Received: September 8, 2021. Revised: November 2, 2021. Accepted: November 19, 2021
© The Author(s) 2021. Published by Oxford University Press on behalf of European Crohn’s and Colitis Organisation. All rights reserved. For permissions,
please email: journals.permissions@oup.com
NAFLD in IBD; Focus on Biologics 853

and differing pharmacotherapy, the variability of diagnostic nosis and staging of NAFLD] and the retrospective design of
methods for NAFLD documentation [different imaging mo- some studies [Table 1].18–24 In this regard, Gizard et al.3 re-
dalities, non-invasive markers in the absence of histological viewed NAFLD prevalence between 1.5% and 55% among
confirmation, being currently the ‘gold-standard’ for the diag- IBD cases, which should be cautiously interpreted. The lower

Table 1. Prevalence of NAFLD among IBD patients and relative risk factors.

First author [year of Country Study design IBD Diagnostic method NAFLD Main risk factors
publication]a cases of NAFLD [%] of NAFLD

Adams et al [2018] Germany Retrospective cross-sectional 130 MRI 54.6 Underweight [BMI <18.5 kg/

Downloaded from https://academic.oup.com/ecco-jcc/article/16/5/852/6433262 by ECCO Member Access user on 24 June 2022


m2]
Balaban et al [2017] Romania Prospective case series 36 CAP-TE 30.5 CD, history of bowel resec-
tion, steroid use, and longer
disease duration
Bargiggia et al [2003] Italy Prospective case-control 511 US 38.0 None
Bessissow et al [2016] Canada Retrospective case series 321 Non-invasive serum 33.6 IBD activity, duration, and
biomarkers, Hepatic history of surgery
Steatosis Index [HIS] and
FIB-4
Bosch et al [2017] USA Retrospective cross-sectional 123 Histology 26.9 Increased BMI and hyper-
tension
Fousekis et al [2019] Greece Retrospective case series 220 Imagingb 20.0 N/A
Glassner et al [2017] USA Retrospective case-control 421 Imagingb 13.3 Older age, prolonged disease
duration, T2DM, increased
BMI, MetS
Hoffmann et al [2020] Germany Retrospective with two parts: 694 US 46.5 Older age, BMI, disease activ-
a cross-sectional and a lon- ity, bowel resection, endo-
gitudinal scopic activity, azathioprine
use
Kang et al [2020] Korea Retrospective case-control 443 CT 11.1 MetS, sarcopenia,
hyperuricaemia, small bowel
resection
Li et al [2017] China Retrospective case-control 206 Imagingb 10.6 For CD: age, female sex, low-
normal BMI, TGF or UC: low
albumin
Likhitsup et al [2019] USA Retrospective cross-sectional 80 US 54.0 CD activity
Magrì et al [2019] Italy Prospective case-control 178 US, TE 40.4 Male sex, older age, obesity,
high caloric consumption
Mancina et al [2016] Italy Prospective case series 158 US, CAP-TE 70.0 PNPLA3-I148M
McHenry et al [2019] USA Prospective case-control 311 MR-PDFF 38.0 BMI <25 kg/m2
Morsy et al [2012] Egypt Prospective case series 33 US 45.5 N/A
Palumbo et al [2018] Canada Prospective case-control 384 CAP elastography 32.8 Age, increased BMI, TG
Principi et al [2018] Italy Prospective case-control 465 US 28.0 T2DM, MetS, abdominal
circumference >102 cm for
males and >88 cm for females.
Ritaccio et al [2020] USA Retrospective cross-sectional 1672 US 12.4 Increased BMI, CD
Sagami et al [2017] Japan Retrospective case-control 303 US 21.8 Age, prolonged disease dur-
ation, low BMI, ileitis
Schroder et al [2015] Germany Retrospective cross-sectional 259 US 28.2 N/A
Simon et al [2018] USA Prospective case series 462 CT 52.0 Age, T2DM, PNPLA3 geno-
type
Sourianarayanane et al USA Retrospective cross-sectional 928 Imaging╪ 8.2 History of surgery, hyperten-
[2013] sion, obesity, steroid use at
imaging
Whorwell et al [1984] UK Case-control 38 US 11.0 N/A
Yamamoto-Furusho [2010] Mexico Prospective case series 200 US 11.2 N/A
Yen et al [2021] Taiwan Retrospective case series 81 US and CAP-TE 29.6 BMI, age at diagnosis

BMI, body mass index; CAP, controlled attenuation parameter; CD, Crohn’s disease; CT, computerised tomography; IBD, inflammatory bowel disease;
MetS, metabolic syndrome; MRI-PDFF, magnetic resonance imaging-proton density fat fraction; NAFLD, non-alcoholic fatty liver disease; N/A, not
applicable; PSC, primary sclerosing cholangitis; T2DM, type 2 diabetes mellitus; TE, transient elastography; TG, triglycerides; TNF, tumour necrosis factor;
UC, ulcerative colitis; US, ultrasound.
a
Studies are sorted according to the last name of the first author.
b
Various imaging modalities included.
854 A. Papaefthymiou et al.

prevalence of NAFLD was mainly reported in older studies, metabolic and genetic risk factors [Figure 1]. Regarding body
using serological, imaging or histological methods of ques- weight, in one study NAFLD was detected, via magnetic res-
tionable validity. A meta-analysis of 19 relevant studies with onance imaging, in 87.6% of underweight patients with IBD,
5620 cases reported that 27.2% of IBD patients had NAFLD,25 who were also more prone to advanced steatosis [>33% of
which is very close to the global prevalence of NAFLD in the hepatocytes] compared with normal-weight individuals; no fi-
general population [25%].5 However, NAFLD prevalence in brosis was presumed, using non-invasive indices,30 thus prob-
IBD may be underestimated in this meta-analysis that dis- ably implying the necessity of further insults [pathogenetic
played marked heterogeneity [I2 = 98%]. In another relevant ‘hits’] to promote liver damage. This rate is much higher than
meta-analysis of 27 studies, the prevalence of NAFLD among the rate reviewed for lean individuals with NAFLD, being
IBD patients was 32%, i.e., somewhat higher than the general 10–20% in the USA and Europe.31 The presence of active in-
population.26 testinal inflammation, assessed by C-reactive protein [CRP]
When considering studies where NAFLD diagnosis was and platelet count, was a significant contributor to NAFLD

Downloaded from https://academic.oup.com/ecco-jcc/article/16/5/852/6433262 by ECCO Member Access user on 24 June 2022


based on methods of high accuracy, such as controlled at- in patients with low body mass index [BMI].30,32 Notably, pa-
tenuation parameter elastography [CAP-E] and magnetic res- tients with NAFLD and IBD in remission had higher BMI.32
onance proton density fat fraction, the prevalence of NAFLD Obesity and aging represent distinct risk factors of NAFLD in
was higher than in general population [Table 1].8,21,27–29 IBD, similarly to the general population in which the propor-
Further well and specifically designed, large epidemiological tion of NAFLD among obese patients ranges between 30%
studies of NAFLD,with cutting edge diagnostic methods, are and 80%.8,14,19,31,33–36 Upon evaluating the disease activity,
required to clarify the prevalence of NAFLD in IBD. BMI and increasing age are risk factors of NAFLD among
patients with IBD in remission in all studies8,34,36–38 but one.33
2.2. Risk factors of NAFLD in IBD Furthermore, sarcopenia has been proposed as an additional
Observational studies have evaluated potential risk factors of independent predisposing factor for NAFLD in IBD patients,
NAFLD in IBD, which may be subdivided into those related after adjustment for other potential cofounders including
with IBD and those not specifically related with IBD, e.g., BMI, T2DM, and MetS.39 The above considered, it seems that

IBD non-specific IBD specific

Overweight Aging
Active Sarcopenia
inflammation

Lack of physical Diet


activity

Bowel IBD
resection duration
T2DM Hypertriglyceridemia

PNPLA3 Anti-integrins Corticosteroids


Hypertension variants

Figure 1. Risk factors associated with NAFLD in patients with IBD. Risk factors are subdivided into those specific and not specific for IBD. Regarding
IBD-specific ones: active and long-term disease, especially CD, small bowel resection, sarcopenia, and some medications have accounted for NAFLD.
IBD non-specific risk factors are common in the general population, including dietary habits, lack of physical activity, obesity, T2DM, dyslipidaemia,
arterial hypertension, aging, and genetic predisposition. Clip arts were obtained by Servier Medical Art [https://smart.servier.com]; no permission is
required. IBD, inflammatory bowel disease; NAFLD, nonalcoholic fatty liver disease; CD, Crohn’s disease; PNPLA, patatin-like phospholipase domain-
containing protein; T2DM, type 2 diabetes mellitus.
NAFLD in IBD; Focus on Biologics 855

sarcopenic obesity may emerge as a predisposing factor for Beyond the classic cytokines, adipokines seem to partici-
NAFLD in the IBD setting,40 as it constitutes a contributor pate in the pathogenesis of chronic inflammatory diseases.63
towards NAFLD in non-IBD populations.41 The most extensively studied adipokines are adiponectin and
Malfunctioning intestinal mucosal barrier has been trad- leptin. Adiponectin acts as an anti-inflammatory, insulin-
itionally associated with NAFLD.42 In this regard, active sensitising and anti-steatotic mediator; 64 it antagonises TNF
IBD-related gut permeability has been incriminated for and IL-6 and, vice versa, it is downregulated by TNF.64,65 In
NAFLD prevalence of 50–60%.10,20,43 Among patients with IBD, adiponectin was shown to be negatively correlated with
active CD, NAFLD rates rise in parallel with CRP44 or with the severity of the disease, although both preclinical and clin-
glucocorticosteroid treatment at the time of NAFLD diagno- ical studies showed controversial results regarding its serum
sis.19,28 Further IBD-associated variables regarded as NAFLD and adipose tissue concentrations.66 Leptin, the prototype
risk factors include disease duration and prior relevant sur- adipokine, shown to be implicated in the pathogenesis of
gery.10,14,19,28,33,36,39 More specifically, a mean disease duration NAFLD,67 seems to increase in the serum and the mesenteric

Downloaded from https://academic.oup.com/ecco-jcc/article/16/5/852/6433262 by ECCO Member Access user on 24 June 2022


of 9–20 years and small bowel resections in patients with adipose tissue of IBD patients, thus contributing to inflam-
CD were independently associated with NAFLD, thus pos- mation, IR, and possibly the NAFLD and cardiovascular
sibly implying an impact of chronic alterations of intestinal burden of these patients.44 Additionally apelin, a peptide as-
pathophysiology, such as small bowel bacterial overgrowth sociated with hepatic fibrosis, has been isolated in the intes-
[SIBO], in NAFLD.10,14,19,28,33,36,39,45 A case-control study has tinal mucosa of IBD patients and in the mesenteric adipose
demonstrated that severe forms of IBD with history of ileal tissue of those with CD, thus comprising another potential
or ileo-colonic resection or total procto-colectomy, frequent linkage between gut inflammation and NAFLD progression.68
relapses [more than one per year] necessitating courses of Apelin may promote liver fibrosis via the extracellular signal-
glucocorticosteroids, and extended intestinal involvement regulated kinase signalling pathway.68
[more than 100 cm for CD or at least left-sided UC] were as- Integrins, transmembrane proteins involved in cell-cell and
sociated with advanced steatosis.46 cell-extracellular matrix interactions, have also been recog-
Moreover, some studies have reported that MetS,14,37,39,47,48 or nised as key molecules in chronic inflammatory and fibrotic
its components such as T2DM,14,37,49 high triglycerides,8,32 arter- conditions, thus constituting rational pharmaceutical targets.
ial hypertentsion19,50 or great waist circumference,37 were asso- The most representative example is β7-integrin, which binds to
ciated with NAFLD. Moreover, a recent meta-analysis showed the mucosal addressin cell adhesion molecule [MAdCAM]-1
that disease duration [16.5–20.0 years], history of IBD-related of intestinal epithelium to facilitate leukocyte migration in
surgery, and methotrexate treatment were IBD-specific risk cases of IBD.69 Although MAdCAM-1 has been considered
factors of NAFLD, along with non-specific risk factors such as a bowel-specific molecule, it is additionally over-expressed
as older age, MetS, its relevant components (T2DM, insulin in chronic liver diseases and may promote fibrosis through
resistance [IR], obesity, arterial hypertension) and CKD.25 The inducing remodelling of the extracellular matrix.70–72
described variability of risk factors strengthens the rationale
of different pathogenetic pillars in IBD-associated NAFLD.40 It 2.4. The role of gut microbiota
seems that in patients with more severe IBD or in those with ac- The symbiosis of host and gut microbiota is disrupted in
tive disease, IBD-specific risk factors may contribute more than chronic inflammatory conditions, such as IBD, thus further
non-specific ones in the pathogenesis of NAFLD, whereas in burdening the inflammatory load locally and systemically.73–75
patients with milder IBD or in those in remission, non-specific Additionally, patients with CD are vulnerable to SIBO, which
risk factors may contribute more than the IBD-specific ones. has also been documented as a distinct risk factor of IR, MetS,
In line, IBD patients in remission were shown to be prone and NAFLD.76–78 Specifically, the disruption of the integrity of
to hypercaloric diets which favour NAFLD.34 However, this intestinal epithelium is supposed to be an early event in IBD
speculation remains to be definitely demonstrated. pathogenesis, permitting bacteria and their products to pene-
trate via a barrier leak, resulting in aberrant immune response
2.3. The role of inflammation and inflammation.79 The imbalance between Bacteroidetes
The inflammatory load, its fluctuations and chronicity, deter- and Firmicutes, to the side of Firmicutes, facilitates the over-
mine the natural history and manifestations of IBD, including growth of abnormal colonies which express mediators of IR
NAFLD. The associated chronic over-expression and system- [lipopolysaccharides] that contribute to local inflammation
atic release of cytokines triggers IR, a key underlying mechan- and possibly to hepatic inflammation through the gut-liver
ism of MetS and NAFLD.51 Cytokines activate an intrahepatic axis.77,78 The intestinal inflammation and the subsequent oxi-
cascade, which facilitates NAFLD development and progres- dative stress disrupt the mucosal tight junctions, thereby al-
sion.52 In this respect, colonic inflammation has been found to lowing bacterial translocation to the liver through the portal
aggravate NASH and fibrosis, thus implying a potential gut- circulation, accompanied by pathogen-associated molecu-
liver axis.53,54 A well-studied cytokine in IBD is tumour necro- lar patterns, danger-associated molecular patterns, reactive
sis factor [TNF], which is also associated with the severity of oxygen species, and Toll-like receptor [TLR]4 and TLR9
NAFLD.55 By acting on its receptor, TNF receptor [TNFR]-I, agonists.74,77,80 As a consequence, an endogenous immune re-
TNF impairs insulin signalling, thus inducing inflammation sponse is induced in the hepatic parenchyma, predisposing to
and IR.56 TNF may also participate in the progression of NAFLD development.74,81
NAFL to NASH and possibly liver fibrosis, by perpetuating In line, preclinical studies investigated potential implica-
the inflammatory process and inducing the local proliferation tions of regulatory molecules, inflammasomes, to dysbiosis-
and activation of the hepatic stellate cells [HSCs].51,55,57–61 related IBD and NAFLD. Common inflammasomes, such
Therefore it has been proposed that active inflammation, pos- as nucleotide-binding oligomerisation domain-like receptor
sibly orchestrated by TNF, may be a key driver of NAFLD protein [NLRP]3 and NLRP6, regulate immune response via
development even in lean IBD patients.62 the expression of IL-1b and IL-18 and serve as modulators
856 A. Papaefthymiou et al.

of normal microbiota, whereas stereotactic abnormalities in activity could be proposed to support their beneficial role.
inflammasomes advocate IBD development through bacter- However, disease remission predisposes to increased nutri-
ial overgrowth.82–84 Other studies revealed that NLRP3 and tional intake and improvement in the absorption of nutrients,
NLRP6 activation improved NAFLD and MetS parameters in leading to obesity, MetS, and NAFLD. The main relevant pre-
rodent models, whereas knockdown of inflammasomes made clinical and clinical data are presented below. Clinical data
mice prone to obesity and NAFLD.85,86 Nevertheless, the exact are also summarised in Table 2, in which it is apparent that
pathophysiological pathways implicated in the inflammasome- there are only observational studies; importantly, the effect
microbiota–NAFLD interaction have not been elucidated. The of biologics on NAFLD was a secondary aim in most cases
dynamic relationships of gut microbiota and the dysbiosis ob- and should carefully interpreted. There are also two ongoing
served in patients with IBD warrant further evaluation to elu- studies for NAFLD in patients with IBD, which are presented
cidate whether changes in the composition of the symbiotic in Table 3. Moreover there are some reports on biologic-
microbiome contribute to the pathogenesis of IBD and its re- induced liver injury, which should be taken into account

Downloaded from https://academic.oup.com/ecco-jcc/article/16/5/852/6433262 by ECCO Member Access user on 24 June 2022


lated NAFLD, which may have therapeutic perspectives. A syn- when evaluating NAFLD.88,89 More specifically, anti-TNF–in-
opsis of all the above considerations is illustrated in Figure 2. duced liver injury has been rarely diagnosed during the in-
duction period, associated with autoimmunity and significant
increase of anti-nuclear antibodies and IgG.89 In such cases,
3. Biologics: Their Potential Effect on NAFLD elevated LFTs are usually normalised after biologic cessation
Τhere are currently conflicting data on the effect of biologics and/or after occasional glucocorticosteroid administration.89
on IBD-associated NAFLD. A relative meta-analysis of seven Vedolizumab, when compared with placebo, was not found
studies, with high heterogeneity [I2 >55%], revealed no asso- to trigger liver injury, albeit isolated cases have been also de-
ciation between IBD pharmaceutical choices and NAFLD.87 scribed.88,90 Awareness of biologic-induced liver injury is im-
Nonetheless, based on the aforementioned rationale and the portant to avoid misdiagnosing NAFLD in rare patients with
proposed mechanisms, decrease in inflammation and disease IBD and high LFTs on biologics.

MAdCAM NAFLD
expression
IR

Downregulation of insulin
HSC signaling pathways
stimulation

Autophagy

TNFα
Gut-liver axis

Adiponectin
ROS LPS
Leptin
TNFα Dysbiosis
Apelin NLRP3/6
Resistin

TNFα

Tight junctions
disruption

PAMPs LPS

ROS
Portal
IBD circulation

Figure 2. The main pathophysiological pathways of IBD related NAFLD. The increased inflammatory load in intestinal mucosa disrupts intracellular tight
junctions, thus allowing the translocation of mediators of inflammation and other molecules in the portal circulation, through which they may affect the
liver. Pro-inflammatory cytokines, products of bacterial dysbiosis, and reactive oxygen species migrate to the hepatic parenchyma, thus triggering IR,
inflammation, and possibly fibrogenesis. Clip arts were obtained by Servier Medical Art [https://smart.servier.com]; no permission is required. HSC,
hepatic stellate cells; IBD, inflammatory bowel disease; IR, insulin resistance; LPS, lipopolysaccharides; MAd-CAM, mucosal addressin cell adhesion
molecule; NAFLD, nonalcoholic fatty liver disease; NLRP, nucleotide-binding oligo-merisation domain-like receptor protein; PAMPs, pathogen-associated
molecular patterns; ROS, reactive oxygen species; TNF, tumour necrosis factor.
NAFLD in IBD; Focus on Biologics 857

Table 2. Clinical studies evaluating the potential effect of biologic agents on NAFLD in patients with IBD

First author [year of Study design Βiologic agent[s] OR or RR 95% CI p-value


publication]a

Bessissow et al [2016] Retrospective case series Anti-TNF 1.69 0.99-2.90 0.056


Glassner et al [2017] Retrospective case-control Anti-TNF, anti-integrin, 0.50 0.20-0.90 0.100
anti-interleukin 12/23
Hoffmann et al [2020] Retrospective with two parts: a cross-sectional and Overall 1.01 0.85-1.17 0.959
a longitudinal
Adalimumab 1.09 0.93-1.25 0.709
Infliximab 1.64 1.48-1.79 0.135

Downloaded from https://academic.oup.com/ecco-jcc/article/16/5/852/6433262 by ECCO Member Access user on 24 June 2022


Golimumab 1.34 1.19-1.50 0.874
Vedolzumab 1.40 1.24-1.56 0.523
Ustekinumab 1.13 0.01-2.25 0.833
Kang et al [2020] Retrospective case-control Not specifically defined 1.07 0.50-2.12 0.856
Magrì et al [2019] Prospective case-control Not specifically defined 0.78 0.42-1.43 0.410
Palumbo et al [2018] Prospective case-control Anti-TNF 0.78 0.50-1.22 0.279
Sagami et al [2017] Retrospective case-control Anti-TNF 1.26 0.71-2.23 0.789
Sourianarayanane et al [2013] Retrospective cross-sectional Anti-TNF 0.63 0.31-1.27 0.194
Yen et al [2021] Retrospective case series Anti-TNF 1.43 0.54-3.81 0.476
Anti-integrin 0.45 0.05–4.09 0.480

IBD, inflammatory bowel disease; NAFLD, non-alcoholic fatty liver disease; OR, odds ratio; RR, relative risk; TNF, tumour necrosis factor; CI, confidence
interval.
a
Studies are sorted according to the last name of the first author.

Table 3. Registered ongoing trials on NAFLD in patients with IBD.

ID, registration date [DD/MM/ Title Design Patients Duration Primary outcome[s]
YYYY]

NCT03760172, 30/11/2018 Immunomediated Non-alcoholic Observational cohort 7300 3 Years Prevalence of NAFLD
SteaTohepatitis; Prevalence and in patients with
Characterization. INSTInCT Study immune-mediated
inflammatory disease
including IBD
NCT04328259, 31/03/2021 Prevalance of Non-Alcoholic Fatty Observational cohort 30 2 years Prevalance of
Liver Diseases in Patient With In- non-alcoholic fatty liver
flammatory Bowel Diseases Attend- in inflammatory bowel
ing Assiut University Hospitals disease participants

IBD, inflammatory bowel disease; NAFLD, non-alcoholic fatty liver disease.

3.1. Anti-TNF agents inflammation, through reduction of intrahepatic expression


The initially introduced biologics in IBD were anti-TNF of TNF and subsequently decreased activation of the Jun
agents and to date they represent a rational choice for the N-terminal kinase [JNK] pathway, resulting in downregulation
management of moderate to severe disease.91,92 Infliximab, a of stearoyl-CoA desaturase, a key enzyme for hepatic trigly-
chimeric monoclonic antibody, was the prototype anti-TNF ceride accumulation.97–99 Furthermore, the hepatic expression
agent used in IBD, followed by adalimumab, golimumab, and of TNF was shown to impede the mitochondrial respiratory
certolizumab pegol.93–96 The amelioration of disease activ- chain and to trigger steatosis, which were resolved after anti-
ity generated the hypothesis that these agents could improve TNF infusion.100 Other authors also investigated the impact
NAFLD at least, through their effect on the inflammation of of anti-TNF on intrahepatic insulin activity by intervening in
both the bowel and the liver and possibly through the de- the Janus kinase [JAK]2/signal transducer and activator of
crease in glucocorticosteroid need.66 Sarcopenia, which is also transcription protein [STAT]3 pathway, which interconnects
related to NAFLD, may be also diminished, since TNF has IR with inflammation;98,101 short-term intraperitoneal treat-
a catabolic role in the skeletal muscle.20 Nevertheless, any ment with infliximab decreased the hepatic inflammatory
beneficial effect of anti-TNF on the intestinal mucosa may load and NAFLD.98 Furthermore it was shown that a 12-week
be accompanied by a ‘rebound’ of weight gain with increased blockage of TNF, in a TNF-knockdown mouse line, decreased
visceral adiposity.20 IR and improved hepatic histology via improving lipid me-
Experimental studies, mainly from obese rodents treated tabolism, inflammatory cytokine expression, and fibrogenic
with anti-TNF agents, have documented improvement factors.102 Moreover by treating high-fat diet models with
in LFTs and, most importantly, in hepatic steatosis and infliximab, visceral fat composition was reduced103 and resistin
858 A. Papaefthymiou et al.

expression was downregulated, thus preventing the develop- ivity in the intestinal mucosa, α4β7 integrin was shown to af-
ment of obesity, T2DM, and NAFLD.104 Notably, the inflam- fect other tissues, thus possibly resulting in the progression of
mation of NASH seems to be improved more effectively than NASH and hepatic fibrosis, probably via facilitating the mi-
steatosis after TNF blockage, probably because TNF acts gration of inflammatory cells through the gut-liver axis.120,121
primarily as an early pro-inflammatory cytokine.99,105 Finally, The first liver disease shown to be implicated by
an orally administered anti-TNF fusion protein, PRX-106, α4β7/MAdCAM-1 treatment was primary scleros-
showed promising results by improving serum and hepatic ing cholangitis, showing α4β7-positive T lymphocytes in
triglycerides, LFTs, and IR.106 These preclinical data provided immunohistochemical analysis.122 Two years later, other au-
a promising background for IBD-associated NAFLD manage- thors documented that, after inhibiting the expression or
ment which, however, necessitates clinical confirmation. functionality of α4β7/MAdCAM in IBD, the hepatic adhe-
Initial clinical data were based on the metabolic effects sion of α4β7 positive T lymphocytes was also impaired, thus
of anti-TNF agents in patients with chronic inflammatory implying a possible role of anti-integrins to manage liver dis-

Downloaded from https://academic.oup.com/ecco-jcc/article/16/5/852/6433262 by ECCO Member Access user on 24 June 2022


diseases, such as rheumatoid or psoriatic arthritis.107 In this eases.121 In this regard, data from mouse models indicated a
regard, a study of 48 psoriatic patients receiving anti-TNF potentially protective effect of α4β7/MAdCAM-1 activation
agents showed that the progression of hepatic steatosis was in T2DM and liver injury.123–125 Nevertheless, a more de-
halted 12 months later, even if the disease activity was min- tailed study on NAFLD showed potentially opposing roles
imally reduced.108 Liver stiffness was also lower in psoriatic for MAdCAM-1 and β7-integrin in NASH.70 More specific-
patients treated with ant-TNF agent, implying an antifibrotic ally, the absence of MAdCAM-1 expression seems to play a
effect.109 Infliximab or adalimumab exerted beneficial effects protective role against hepatic steatosis.70 In this regard, the
on IR and central obesity, leading to a subsequent improve- role of the α4β7 complex, which activates MAdCAM-1 in
ment in MetS-related components such as NAFLD, and in the liver, remains conflicting since β7 integrin knockdown
cardiovascular risk.110–112 Importantly, studies investigating mice were, unexpectedly, prone to NASH development.70
the impact of anti-TNF agents on dyslipidaemia showed an These complicated and largely unresolved interactions have
increase in serum high-density lipoprotein-cholesterol with- triggered an ongoing debate on the potential role of α4β7
out affecting other lipoproteins, whereas a potentially adverse complex and MAdCAM-1 in the gut-liver axis, which war-
effect was recorded on triglyceride concentrations.113–116 rant further research.
Limited retrospective clinical studies investigated the effect Despite the scarcity of relevant clinical data, a recent
of anti-TNF agents on NAFLD in patients with IBD, albeit as retrospective cohort study, using CAP-E, documented that
a confounder. Infliximab, 14 weeks after the first dose, did not NAFLD patients with IBD had higher rates of treatment
restore IR and led to higher BMI in IBD patients with clinical with vedolizumab [16.7%] compared with those without
response.117 In longer-term studies, anti-TNF achieving remis- NAFLD [1.8%]; however, vedolizumab treatment was neither
sion was also associated with higher probability of weight the primary aim of the study nor independently associated
gain, especially in patients with CD.20 These results warrant with NAFLD after adjustment for potential confounders.38
further research, since the subsequent weight gain may at- Moreover, a study reviewing the safety of vedolizumab con-
tenuate any beneficial effect of anti-TNF agents on NAFLD. cluded that, in patients with IBD experiencing hepatic adverse
Most studies in IBD patients do not show an association be- events, NAFLD was the most common diagnosis.90 Thus,
tween the treatment with biologics and NAFLD rates and/or until further studies elucidate this topic, anti-integrin treat-
severity.8,10,14,21,33,34,36,38,39 ment is not recommended in patients with IBD and NAFLD.
In one retrospective study,19 patients with IBD not on anti-
TNF had double the rate of NAFLD compared with those 3.3. Anti-IL-12/23 agents
on anti-TNF, albeit without being statistically significant.19 In IL-23 participates in autoimmunity pathways by triggering
another study, anti-TNF treatment was the only factor that the differentiation of T helper-17 lymphocytes. In this re-
was inversely associated with NAFLD severity in patients gard ustekinumab, an anti-IL12/23 monoclonal antibody
with IBD, as implied from LFTs.46 On the contrary, other au- directed against the p40 protein subunit of IL-23 and IL-12,
thors43 reported a high rate of NAFLD in patients on anti- had been initially introduced in the treatment of psoriasis.126
TNF [54%]; however, in this study there was not a control Subsequently, ustekinumab was approved for CD in 2016
group.43 Of note, in the same study, NAFLD patients had in- and then for UC.127,128 However, due to its short-term clinical
dependently higher CD activity than those without NAFLD.43 use, limited data exist on its effect on NAFLD in patients with
These findings warrant the need of further prospective studies IBD.14,33
in carefully selected patients with IBD and a primary endpoint In one retrospective study, ustekinumab treatment, used as
of the effect of anti-TNF on NAFLD parameters, especially a potential confounder, was not associated with NAFLD.33
those related to hepatic inflammation and fibrosis; in this re- Nevertheless, data from its administration in extra-intestinal
gard, studies with paired liver biopsies may be considered to diseases indicated that treatment with anti-interleukin 12/23
be the optimal design, but due to their invasive nature, studies seems to positively affect lipid metabolism, through its effect
with parameters of elastography and/or non-invasive indices on leptin expression. More specifically ustekinumab induces
of hepatic fibrosis may be acceptable alternatives. leptin, suggesting a potential benefit for MetS and visceral
adiposity.129 Other authors also showed that patients receiv-
3.2. Anti-integrin agents ing anti-IL12/23 do not gain weight, in contrast to those
In 2014, an anti-α4β7 integrin monoclonal antibody, treated with anti-TNF agents.130
vedolizumab, was introduced into clinical practice for pa- However, induction of leptin may have a dual role in
tients with IBD, based on the GEMINI study which targeted NAFLD: leptin was shown to be beneficial in the early
remission and its maintenance for both naïve and non-naïve stages of NAFLD through its anti-steatotic effect, but may
patients.118,119 Despite the initial notion of its absolute select- favour NAFLD progression in the later stages through its
NAFLD in IBD; Focus on Biologics 859

inflammatory and fibrogenic action.67,101 Therefore, until more Author Contributions


data specifically clarify the role of anti-IL12/23 in NAFLD, its
AP designed the study, performed the research, collected and
use in patients with IBD and NAFLD should be carefully con-
analysed the data, and wrote the paper; SP, AG, MD, and JK
sidered, especially in cases of NASH and hepatic fibrosis.
further analysed the collected data, drafted and critically re-
vised the manuscript; SAP designed the study, drafted and
4. Closing Remarks critically revised the manuscript. All the authors approved the
final version of this article.
NAFLD seems to co-exist in a high proportion of patients
with IBD and to follow two distinct forms, most likely reflect-
ing different predominant pathophysiological mechanisms
during IBD relapses and remissions. Intestinal inflammation
References
facilitates the dysbiosis of microbiota, mucosal disruption, 1. Burisch J, Kiudelis G, Kupcinskas L, et al.; Epi-IBD group. Natural

Downloaded from https://academic.oup.com/ecco-jcc/article/16/5/852/6433262 by ECCO Member Access user on 24 June 2022


release of pro-inflammatory cytokines, and translocation of disease course of Crohn’s disease during the first 5 years after diag-
nosis in a European population-based inception cohort: an Epi-IBD
inflammatory cells through the gut-liver axis, thus represent-
study. Gut 2019;68:423–33.
ing the predominant type of NAFLD during disease relapses.
2. Algaba A, Guerra I, Ricart E, et al.; Spanish GETECCU Group
During remissions, high-calorie nutrition and the restored [ENEIDA Project]. Extraintestinal manifestations in patients with
mucosal absorption may contribute to weight gain, thus inflammatory bowel disease: study based on the ENEIDA Registry.
predisposing to dysmetabolism and thus to NAFLD. Due to Dig Dis Sci 2021;66:2014–23.
this special and distinct association between IBD and NAFLD, 3. Gizard E, Ford AC, Bronowicki JP, Peyrin-Biroulet L. Systematic
which includes the classic predisposing factors [dysmetabolic review: the epidemiology of the hepatobiliary manifestations in pa-
ones] and also the disease-specific ones, we may consider the tients with inflammatory bowel disease. Aliment Pharmacol Ther
introduction of a specific NAFLD type which could be named 2014;40:3–15.
IBD-associated fatty liver disease [IBDAFLD]. This may be 4. Restellini S, Chazouillères O, Frossard JL. Hepatic manifestations
of inflammatory bowel diseases. Liver Int 2017;37:475–89.
a subtype of the metabolic dysfunction-associated fatty liver
5. Makri E, Goulas A, Polyzos SA. Epidemiology, pathogenesis, diag-
disease [MAFLD],131 representing an emerging nomenclature,
nosis and emerging treatment of nonalcoholic fatty liver disease.
but also a different definition of the disease despite fruitful Arch Med Res 2021;52:25–37.
criticism by us and other authors.132 6. Fazel Y, Koenig AB, Sayiner M, Goodman ZD, Younossi ZM. Epi-
There is current necessity of a holistic management of pa- demiology and natural history of non-alcoholic fatty liver disease.
tients with IBD. In this regard the effect of biologics, which Metabolism 2016;65:1017–25.
have been largely introduced in the treatment of IBD, on 7. Nguyen DL, Bechtold ML, Jamal MM. National trends and in-
NAFLD should be scrutinised. The relevant scientific lit- patient outcomes of inflammatory bowel disease patients
erature is inconsistent, probably reflecting the diversity of with concomitant chronic liver disease. Scand J Gastroenterol
action of different biologics as well as the lack of studies 2014;49:1091–5.
8. Saroli Palumbo C, Restellini S, Chao CY, et al. Screening for
specifically designed for NAFLD endpoints. The more exten-
nonalcoholic fatty liver disease in inflammatory bowel diseases:
sively studied anti-TNF agents, having been included mostly
a cohort study using transient elastography. Inflamm Bowel Dis
as confounders in relevant studies, showed a neutral or a po- 2019;25:124–33.
tentially beneficial association with NAFLD. On the contrary 9. Polyzos SA, Kountouras J, Mantzoros CS. Obesity and nonalcoholic
the anti-integrin antibody, vedolizumab, may lead to the fatty liver disease: from pathophysiology to therapeutics. Metabol-
progression of NAFLD, based on limited data from not spe- ism 2019;92:82–97.
cifically designed studies. Anti-IL12/23 are even less studied 10. Bessissow T, Le NH, Rollet K, Afif W, Bitton A, Sebastiani G. Inci-
in NAFLD, but should be cautiously considered in patients dence and predictors of nonalcoholic fatty liver disease by serum
with NASH and advanced fibrosis, because they upregulate biomarkers in patients with inflammatory bowel disease. Inflamm
leptin which may have adverse effect in NASH and hepatic Bowel Dis 2016;22:1937–44.
11. Long MD, Crandall WV, Leibowitz IH, et al.; ImproveCareNow
fibrosis. Current data warrant further mechanistic studies
Collaborative for Pediatric IBD. Prevalence and epidemiology of
to enlighten the mechanisms of action of biologics, as well
overweight and obesity in children with inflammatory bowel dis-
as prospective cohort studies and possibly, clinical trials in ease. Inflamm Bowel Dis 2011;17:2162–8.
patients with IBD and concomitant NAFLD. In this way, we 12. Rosen E, Bakshi N, Watters A, Rosen HR, Mehler PS. Hepatic
will be able to select the best biologic agent for each patient complications of anorexia nervosa. Dig Dis Sci 2017;62:2977–81.
with IBD, achieving together a potentially beneficial or at 13. Rautou PE, Cazals-Hatem D, Moreau R, et al. Acute liver cell
least neutral effect on NAFLD [or IBDAFLD]. If a beneficial damage in patients with anorexia nervosa: a possible role of
effect of some biologics is shown in patients with IBD, this starvation-induced hepatocyte autophagy. Gastroenterology
may open a new window for clinical trials of biologics in 2008;135:840–8, 848.e1–3.
NAFLD patients without IBD, as no approved treatment for 14. Glassner K, Malaty HM, Abraham BP. Epidemiology and risk
factors of nonalcoholic fatty liver disease among patients with in-
NAFLD exists.133
flammatory bowel disease. Inflamm Bowel Dis 2017;23:998–1003.
15. Polyzos SA, Kang ES, Boutari C, Rhee EJ, Mantzoros CS. Current
and emerging pharmacological options for the treatment of nonal-
Funding coholic steatohepatitis. Metabolism 2020;111:154203.
There was no grant or financial support for this review. 16. Berg DR, Colombel JF, Ungaro R. The role of early biologic therapy in
inflammatory bowel disease. Inflamm Bowel Dis 2019;25:1896–905.
17. Piovani D, Danese S, Peyrin-Biroulet L, Nikolopoulos GK,
Bonovas S. Systematic review with meta-analysis: biologics and
Conflict of Interest
risk of infection or cancer in elderly patients with inflammatory
No author has any conflict of interest. bowel disease. Aliment Pharmacol Ther 2020;51:820–30.
860 A. Papaefthymiou et al.

18. Chao CY, Battat R, Al Khoury A, Restellini S, Sebastiani G, 38. Yen HH, Su PY, Huang SP, et al. Evaluation of non-alcoholic fatty
Bessissow T. Co-existence of non-alcoholic fatty liver disease and liver disease in patients with inflammatory bowel disease using
inflammatory bowel disease: a review article. World J Gastroenterol controlled attenuation parameter technology: a Taiwanese retro-
2016;22:7727–34. spective cohort study. PLoS One 2021;16:e0252286.
19. Sourianarayanane A, Garg G, Smith TH, Butt MI, McCullough AJ, 39. Kang MK, Kim KO, Kim MC, Park JG, Jang BI. Sarcopenia is a
Shen B. Risk factors of non-alcoholic fatty liver disease in patients new risk factor of nonalcoholic fatty liver disease in patients with
with inflammatory bowel disease. J Crohns Colitis 2013;7:e279– inflammatory bowel disease. Dig Dis 2020;38:507–14.
85. 40. Gibiino G, Sartini A, Gitto S, et al. The other side of malnutrition
20. McGowan CE, Jones P, Long MD, Barritt AS 4th. Changing shape in inflammatory bowel disease [IBD]: non-alcoholic fatty liver dis-
of disease: nonalcoholic fatty liver disease in Crohn’s disease ease. Nutrients 2021;13:2772.
– a case series and review of the literature. Inflamm Bowel Dis 41. Polyzos SA, Margioris AN. Sarcopenic obesity. Hormones
2012;18:49–54. 2018;17:321–31.
21. McHenry S, Sharma Y, Tirath A, et al. Crohn’s disease is associated 42. Miele L, Valenza V, La Torre G, et al. Increased intestinal permea-

Downloaded from https://academic.oup.com/ecco-jcc/article/16/5/852/6433262 by ECCO Member Access user on 24 June 2022


with an increased prevalence of nonalcoholic fatty liver disease: a bility and tight junction alterations in nonalcoholic fatty liver dis-
cross-sectional study using magnetic resonance proton density fat ease. Hepatology 2009;49:1877–87.
fraction mapping. Clin Gastroenterol Hepatol 2019;17:2816–8. 43. Likhitsup A, Dundulis J, Ansari S, et al. High prevalence of
22. Fousekis FS, Katsanos KH, Theopistos VI, et al. Hepatobiliary and non-alcoholic fatty liver disease in patients with inflammatory
pancreatic manifestations in inflammatory bowel diseases: a re- bowel disease receiving anti-tumor necrosis factor therapy. Ann
ferral center study. BMC Gastroenterol 2019;19:48. Gastroenterol 2019;32:463–8.
23. Polyzos SA, Mantzoros CS. Necessity for timely noninvasive diag- 44. Maher JJ, Leon P, Ryan JC. Beyond insulin resistance: innate immu-
nosis of nonalcoholic fatty liver disease. Metabolism 2014;63:161– nity in nonalcoholic steatohepatitis. Hepatology 2008;48:670–8.
7. 45. Wijarnpreecha K, Lou S, Watthanasuntorn K, et al. Small intes-
24. Yamamoto-Furusho JK, Sánchez-Osorio M, Uribe M. Prevalence tinal bacterial overgrowth and nonalcoholic fatty liver disease: a
and factors associated with the presence of abnormal function liver systematic review and meta-analysis. Eur J Gastroenterol Hepatol
tests in patients with ulcerative colitis. Ann Hepatol 2010;9:397– 2020;32:601–8.
401. 46. Sartini A, Gitto S, Bianchini M, et al. Non-alcoholic fatty liver dis-
25. Zou ZY, Shen B, Fan JG. Systematic review with meta-analysis: ease phenotypes in patients with inflammatory bowel disease. Cell
epidemiology of nonalcoholic fatty liver disease in patients with in- Death Dis 2018;9:87.
flammatory bowel disease. Inflamm Bowel Dis 2019;25:1764–72. 47. Carr RM, Patel A, Bownik H, et al. Intestinal inflammation does
26. Lin A, Roth H, Anyane-Yeboa A, Rubin DT, Paul S. Prevalence not predict nonalcoholic fatty liver disease severity in inflammatory
of nonalcoholic fatty liver disease in patients with inflammatory bowel disease patients. Dig Dis Sci 2017;62:1354–61.
bowel disease: a systematic review and meta-analysis. Inflamm 48. Spagnuolo R, Abenavoli L, Corea A, et al. Multifaceted pathogen-
Bowel Dis 2021;27:947–55. esis of liver steatosis in inflammatory bowel disease: a systematic
27. Morsy KH, Hasanain AF. Hepatobiliary disorders among naïve review. Eur Rev Med Pharmacol Sci 2021;25:5818–25.
patients with ulcerative colitis in Upper Egypt. Arab J Gastroenterol 49. Simon TG, Van Der Sloot KWJ, Chin SB, et al. IRGM gene
2012;13:71–6. variants modify the relationship between visceral adipose tissue
28. Balaban DV, Popp A, Robu G, et al. Fatty liver assessment in in- and NAFLD in patients with Crohn’s disease. Inflamm Bowel Dis
flammatory bowel disease patients using controlled attenuation pa- 2018;24:2247–57.
rameter. J Crohns Colitis 2017;11[Suppl_1]:S240–1. 50. Bosch DE, Yeh MM. Primary sclerosing cholangitis is protective
29. Mancina RM, Spagnuolo R, Milano M, et al. PNPLA3 148M against nonalcoholic fatty liver disease in inflammatory bowel dis-
carriers with inflammatory bowel diseases have higher suscepti- ease. Hum Pathol 2017;69:55–62.
bility to hepatic steatosis and higher liver enzymes. Inflamm Bowel 51. Hotamisligil GS. Inflammation and metabolic disorders. Nature
Dis 2016;22:134–40. 2006;444:860–7.
30. Adams LC, Lübbe F, Bressem K, Wagner M, Hamm B, 52. Polyzos SA, Kountouras J, Mantzoros CS. Adipokines in nonalco-
Makowski MR. Non-alcoholic fatty liver disease in underweight holic fatty liver disease. Metabolism 2016;65:1062–79.
patients with inflammatory bowel disease: a case-control study. 53. Coffin CS, Fraser HF, Panaccione R, Ghosh S. Liver diseases associ-
PLoS One 2018;13:e0206450. ated with anti-tumor necrosis factor-alpha [TNF-α] use for inflam-
31. Younossi Z, Anstee QM, Marietti M, et al. Global burden of matory bowel disease. Inflamm Bowel Dis 2011;17:479–84.
NAFLD and NASH: trends, predictions, risk factors and preven- 54. Gäbele E, Dostert K, Hofmann C, et al. DSS induced co-
tion. Nat Rev Gastroenterol Hepatol 2018;15:11–20. litis increases portal LPS levels and enhances hepatic inflam-
32. Li D, Lu C, Yu C. High incidence of non-alcoholic fatty liver disease mation and fibrogenesis in experimental NASH. J Hepatol
in patients with Crohn’s disease but not ulcerative colitis. Int J Clin 2011;55:1391–9.
Exp Pathol 2017;10:10633–9. 55. Potoupni V, Georgiadou M, Chatzigriva E, et al. Circulating
33. Hoffmann P, Jung V, Behnisch R, Gauss A. Prevalence and risk tumor necrosis factor-α levels in non-alcoholic fatty liver disease:
factors of nonalcoholic fatty liver disease in patients with inflam- a systematic review and a meta-analysis. J Gastroenterol Hepatol
matory bowel diseases: a cross-sectional and longitudinal analysis. 2021;36:3002–14.
World J Gastroenterol 2020;26:7367–81. 56. Popa C, Netea MG, van Riel PL, van der Meer JW, Stalenhoef AF.
34. Magrì S, Paduano D, Chicco F, et al. Nonalcoholic fatty liver dis- The role of TNF-alpha in chronic inflammatory conditions, in-
ease in patients with inflammatory bowel disease: beyond the nat- termediary metabolism, and cardiovascular risk. J Lipid Res
ural history. World J Gastroenterol 2019;25:5676–86. 2007;48:751–62.
35. Ritaccio G, Stoleru G, Abutaleb A, et al. Nonalcoholic fatty liver dis- 57. Harrison SA, Kadakia S, Lang KA, Schenker S. Nonalcoholic
ease is common in IBD patients; however progression to hepatic fi- steatohepatitis: what we know in the new millennium. Am J
brosis by noninvasive markers is rare. Dig Dis Sci 2021;66:3186–91. Gastroenterol 2002;97:2714–24.
36. Sagami S, Ueno Y, Tanaka S, et al. Significance of non-alcoholic 58. Ikejima K, Takei Y, Honda H, et al. Leptin receptor-mediated
fatty liver disease in Crohn’s disease: a retrospective cohort study. signaling regulates hepatic fibrogenesis and remodeling of extracel-
Hepatol Res 2017;47:872–81. lular matrix in the rat. Gastroenterology 2002;122:1399–410.
37. Principi M, Iannone A, Losurdo G, et al. Nonalcoholic fatty liver 59. Singh S, Young P, Armstrong AW. An update on psoriasis and met-
disease in inflammatory bowel disease: prevalence and risk factors. abolic syndrome: a meta-analysis of observational studies. PLoS
Inflamm Bowel Dis 2018;24:1589–96. One 2017;12:e0181039.
NAFLD in IBD; Focus on Biologics 861

60. Persico M, Capasso M, Persico E, et al. Suppressor of cytokine microbiota on the development and progression of nonalcoholic
signaling 3 [SOCS3] expression and hepatitis C virus-related steatohepatitis. Eur J Nutr 2018;57:861–76.
chronic hepatitis: insulin resistance and response to antiviral 82. Wood NJ. Microbiota: dysbiosis driven by inflammasome defi-
therapy. Hepatology 2007;46:1009–15. ciency exacerbates hepatic steatosis and governs rate of NAFLD
61. Tomita K, Tamiya G, Ando S, et al. Tumour necrosis factor alpha progression. Nat Rev Gastroenterol Hepatol 2012;9:123.
signalling through activation of Kupffer cells plays an essential 83. Zaki MH, Boyd KL, Vogel P, Kastan MB, Lamkanfi M,
role in liver fibrosis of non-alcoholic steatohepatitis in mice. Gut Kanneganti TD. The NLRP3 inflammasome protects against loss of
2006;55:415–24. epithelial integrity and mortality during experimental colitis. Im-
62. Hotamisligil GS, Peraldi P, Budavari A, Ellis R, White MF, munity 2010;32:379–91.
Spiegelman BM. IRS-1-mediated inhibition of insulin receptor ty- 84. Hirota SA, Ng J, Lueng A, et al. NLRP3 inflammasome plays a key
rosine kinase activity in TNF-alpha- and obesity-induced insulin role in the regulation of intestinal homeostasis. Inflamm Bowel Dis
resistance. Science 1996;271:665–8. 2011;17:1359–72.
63. Polyzos SA, Kountouras J, Mantzoros CS. Adipose tissue, obe- 85. Henao-Mejia J, Elinav E, Jin C, et al. Inflammasome-mediated

Downloaded from https://academic.oup.com/ecco-jcc/article/16/5/852/6433262 by ECCO Member Access user on 24 June 2022


sity and non-alcoholic fatty liver disease. Minerva Endocrinol dysbiosis regulates progression of NAFLD and obesity. Nature
2017;42:92–108. 2012;482:179–85.
64. Polyzos SA, Kountouras J, Zavos C, Tsiaousi E. The role of 86. Pierantonelli I, Rychlicki C, Agostinelli L, et al. Author Correction:
adiponectin in the pathogenesis and treatment of non-alcoholic lack of NLRP3-inflammasome leads to gut-liver axis derangement,
fatty liver disease. Diabetes Obes Metab 2010;12:365–83. gut dysbiosis and a worsened phenotype in a mouse model of
65. Polyzos SA, Kountouras J, Zavos Ch. The multi-hit process and the NAFLD. Sci Rep 2017;7:17568.
antagonistic roles of tumor necrosis factor-alpha and adiponectin 87. Lapumnuaypol K, Kanjanahattakij N, Pisarcik D, Thongprayoon C,
in non alcoholic fatty liver disease. Hippokratia 2009;13:127; au- Wijarnpreecha K, Cheungpasitporn W. Effects of inflammatory
thor reply 128. bowel disease treatment on the risk of nonalcoholic fatty liver dis-
66. Gonçalves P, Magro F, Martel F. Metabolic inflammation in inflam- ease: a meta-analysis. Eur J Gastroenterol Hepatol 2018;30:854–
matory bowel disease: crosstalk between adipose tissue and bowel. 60.
Inflamm Bowel Dis 2015;21:453–67. 88. Honap S, Sticova E, Theocharidou E, et al. Vedolizumab-associated
67. Polyzos SA, Kountouras J, Mantzoros CS. Leptin in nonalcoholic drug-induced liver injury: a case series. Inflamm Bowel Dis
fatty liver disease: a narrative review. Metabolism 2015;64:60–78. 2021;27:e32–4.
68. Wang Y, Song J, Bian H, et al. Apelin promotes hepatic fi- 89. Björnsson HK, Gudbjornsson B, Björnsson ES. Infliximab-induced
brosis through ERK signaling in LX-2 cells. Mol Cell Biochem liver injury: clinical phenotypes, autoimmunity and the role of cor-
2019;460:205–15. ticosteroid treatment. J Hepatol 2021.
69. Tiisala S, Paavonen T, Renkonen R. Alpha E beta 7 and alpha 4 beta 90. Colombel JF, Sands BE, Rutgeerts P, et al. The safety of vedolizumab
7 integrins associated with intraepithelial and mucosal homing, are for ulcerative colitis and Crohn’s disease. Gut 2016;66:839–51.
expressed on macrophages. Eur J Immunol 1995;25:411–7. 91. Gomollón F, Dignass A, Annese V, et al.; ECCO. Third European
70. Drescher HK, Schippers A, Clahsen T, et al. β7-Integrin and evidence-based consensus on the diagnosis and management of
MAdCAM-1 play opposing roles during the development of non- Crohn’s disease 2016. Part 1: diagnosis and medical management.
alcoholic steatohepatitis. J Hepatol 2017;66:1251–64. J Crohns Colitis 2017;11:3–25.
71. Schippers A, Hübel J, Heymann F, et al. MAdCAM-1/α4β7 92. Harbord M, Eliakim R, Bettenworth D, et al. Third European evi-
integrin-mediated lymphocyte/endothelium interactions exacerbate dence-based consensus on diagnosis and management of ulcerative
acute immune-mediated hepatitis in mice. Cell Mol Gastroenterol colitis. Part 2: current management. J Crohns Colitis 2017;11:769–
Hepatol 2021;11:1227–50.e1. 84.
72. Ester C, Cerban R, Iacob S, et al. The role of beta-7 integrin and 93. Sandborn WJ, Feagan BG, Marano C, et al.; PURSUIT-Maintenance
carbonic anhydrase IX in predicting the occurrence of de novo non- Study Group. Subcutaneous golimumab maintains clinical response
alcoholic fatty liver disease in liver transplant recipients. Chirurgia in patients with moderate-to-severe ulcerative colitis. Gastroenter-
2018;113:534–41. ology 2014;146:96–109.e1.
73. Torres J, Palmela C, Brito H, et al. The gut microbiota, bile acids and their 94. Hanauer SB, Sandborn WJ, Rutgeerts P, et al. Human anti-tumor
correlation in primary sclerosing cholangitis associated with inflamma- necrosis factor monoclonal antibody [adalimumab] in Crohn’s dis-
tory bowel disease. United European Gastroenterol J 2018;6:112–22. ease: the CLASSIC-I trial. Gastroenterology 2006;130:323–33;
74. Acharya C, Sahingur SE, Bajaj JS. Microbiota, cirrhosis, and the quiz 591.
emerging oral-gut-liver axis. JCI Insight 2017;2. 95. Hanauer SB, Feagan BG, Lichtenstein GR, et al.; ACCENT I Study
75. Nguyen NH, Ohno-Machado L, Sandborn WJ, Singh S. Obesity is Group. Maintenance infliximab for Crohn’s disease: the ACCENT
independently associated with higher annual burden and costs of I randomised trial. Lancet 2002;359:1541–9.
hospitalization in patients with inflammatory bowel diseases. Clin 96. Rawla P, Sunkara T, Raj JP. Role of biologics and biosimilars in in-
Gastroenterol Hepatol 2019;17:709–18.e7. flammatory bowel disease: current trends and future perspectives. J
76. Diamant M, Blaak EE, de Vos WM. Do nutrient-gut-microbiota Inflamm Res 2018;11:215–26.
interactions play a role in human obesity, insulin resistance and 97. Li Z, Yang S, Lin H, et al. Probiotics and antibodies to TNF inhibit
type 2 diabetes? Obes Rev 2011;12:272–81. inflammatory activity and improve nonalcoholic fatty liver disease.
77. Abu-Shanab A, Quigley EM. The role of the gut microbiota in Hepatology 2003;37:343–50.
nonalcoholic fatty liver disease. Nat Rev Gastroenterol Hepatol 98. Barbuio R, Milanski M, Bertolo MB, Saad MJ, Velloso LA. Infliximab
2010;7:691–701. reverses steatosis and improves insulin signal transduction in liver of
78. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, rats fed a high-fat diet. J Endocrinol 2007;194:539–50.
Gordon JI. An obesity-associated gut microbiome with increased 99. Koca SS, Bahcecioglu IH, Poyrazoglu OK, Ozercan IH, Sahin K,
capacity for energy harvest. Nature 2006;444:1027–31. Ustundag B. The treatment with antibody of TNF-alpha reduces
79. Chelakkot C, Choi Y, Kim DK, et al. Akkermansia muciniphila- the inflammation, necrosis and fibrosis in the non-alcoholic
derived extracellular vesicles influence gut permeability through steatohepatitis induced by methionine- and choline-deficient diet.
the regulation of tight junctions. Exp Mol Med 2018;50:e450. Inflammation 2008;31:91–8.
80. Bischoff SC, Barbara G, Buurman W, et al. Intestinal perme- 100. García-Ruiz I, Rodríguez-Juan C, Díaz-Sanjuan T, et al. Uric acid
ability – a new target for disease prevention and therapy. BMC and anti-TNF antibody improve mitochondrial dysfunction in ob/
Gastroenterol 2014;14:189. ob mice. Hepatology 2006;44:581–91.
81. de Faria Ghetti F, Oliveira DG, de Oliveira JM, 101. Polyzos SA, Kountouras J, Zavos C, Deretzi G. The potential ad-
de Castro Ferreira LEVV, Cesar DE, Moreira APB. Influence of gut verse role of leptin resistance in nonalcoholic fatty liver disease:
862 A. Papaefthymiou et al.

a hypothesis based on critical review of the literature. J Clin levels and insulin resistance in patients with inflammatory bowel
Gastroenterol 2011;45:50–4. disease. Eur J Gastroenterol Hepatol 2009;21:283–8.
102. Kakino S, Ohki T, Nakayama H, et al. Pivotal role of TNF-α in the 118. Feagan BG, Rutgeerts P, Sands BE, et al.; GEMINI 1 Study Group.
development and progression of nonalcoholic fatty liver disease in Vedolizumab as induction and maintenance therapy for ulcerative
a murine model. Horm Metab Res 2018;50:80–7. colitis. N Engl J Med 2013;369:699–710.
103. Vignozzi L, Filippi S, Comeglio P, et al. Nonalcoholic steatohepatitis 119. Sands BE, Feagan BG, Rutgeerts P, et al. Effects of vedolizumab in-
as a novel player in metabolic syndrome-induced erectile dysfunc- duction therapy for patients with Crohn’s disease in whom tumor
tion: an experimental study in the rabbit. Mol Cell Endocrinol necrosis factor antagonist treatment failed. Gastroenterology
2014;384:143–54. 2014;147:618–27.e3.
104. Campanati A, Ganzetti G, Giuliodori K, et al. Serum levels of 120. Adams DH, Eksteen B, Curbishley SM. Immunology of the gut
adipocytokines in psoriasis patients receiving tumor necrosis and liver: a love/hate relationship. Gut 2008;57:838–48.
factor-α inhibitors: results of a retrospective analysis. Int J 121. Grant AJ, Lalor PF, Hübscher SG, Briskin M, Adams DH.
Dermatol 2015;54:839–45. MAdCAM-1 expressed in chronic inflammatory liver disease

Downloaded from https://academic.oup.com/ecco-jcc/article/16/5/852/6433262 by ECCO Member Access user on 24 June 2022


105. Iimuro Y, Gallucci RM, Luster MI, Kono H, Thurman RG. supports mucosal lymphocyte adhesion to hepatic endothelium
Antibodies to tumor necrosis factor alfa attenuate hepatic ne- [MAdCAM-1 in chronic inflammatory liver disease]. Hepatology
crosis and inflammation caused by chronic exposure to ethanol in 2001;33:1065–72.
the rat. Hepatology 1997;26:1530–7. 122. Ponsioen CY, Kuiper H, Ten Kate FJ, van Milligen de Wit M,
106. Ilan Y, Ben Ya’acov A, Shabbat Y, Gingis-Velitski S, Almon E, van Deventer SJ, Tytgat GN. Immunohistochemical analysis of in-
Shaaltiel Y. Oral administration of a non-absorbable plant flammation in primary sclerosing cholangitis. Eur J Gastroenterol
cell-expressed recombinant anti-TNF fusion protein induces Hepatol 1999;11:769–74.
immunomodulatory effects and alleviates nonalcoholic 123. Yang XD, Sytwu HK, McDevitt HO, Michie SA. Involvement of
steatohepatitis. World J Gastroenterol 2016;22:8760–9. beta 7 integrin and mucosal addressin cell adhesion molecule-1
107. Jones RE, Moreland LW. Tumor necrosis factor inhibitors for [MAdCAM-1] in the development of diabetes in obese diabetic
rheumatoid arthritis. Bull Rheum Dis 1999;48:1–4. mice. Diabetes 1997;46:1542–7.
108. Di Minno MN, Iervolino S, Peluso R, et al.; CaRRDS Study 124. Xu B, Cook RE, Michie SA. Alpha4beta7 integrin/MAdCAM-1 ad-
Group. Hepatic steatosis and disease activity in subjects with hesion pathway is crucial for B cell migration into pancreatic lymph
psoriatic arthritis receiving tumor necrosis factor-α blockers. J nodes in nonobese diabetic mice. J Autoimmun 2010;35:124–9.
Rheumatol 2012;39:1042–6. 125. Wolf D, Hallmann R, Sass G, et al. TNF-alpha-induced expres-
109. Seitz M, Reichenbach S, Möller B, Zwahlen M, Villiger PM, sion of adhesion molecules in the liver is under the control of
Dufour JF. Hepatoprotective effect of tumour necrosis factor TNFR1 – relevance for concanavalin A-induced hepatitis. J
alpha blockade in psoriatic arthritis: a cross-sectional study. Ann Immunol 2001;166:1300–7.
Rheum Dis 2010;69:1148–50. 126. Strober BE, Bissonnette R, Fiorentino D, et al. Comparative ef-
110. Maruotti N, d’Onofrio F, Cantatore FP. Metabolic syndrome and fectiveness of biologic agents for the treatment of psoriasis in
chronic arthritis: effects of anti-TNF-α therapy. Clin Exp Med a real-world setting: results from a large, prospective, observa-
2015;15:433–8. tional study [Psoriasis Longitudinal Assessment and Registry
111. Costa L, Caso F, Atteno M, et al. Impact of 24-month treatment (PSOLAR)]. J Am Acad Dermatol 2016;74:851–61.e4.
with etanercept, adalimumab, or methotrexate on metabolic syn- 127. Feagan BG, Sandborn WJ, Gasink C, et al.; UNITI–IM-UNITI
drome components in a cohort of 210 psoriatic arthritis patients. Study Group. Ustekinumab as induction and maintenance therapy
Clin Rheumatol 2014;33:833–9. for Crohn’s disease. N Engl J Med 2016;375:1946–60.
112. Barnabe C, Martin BJ, Ghali WA. Systematic review and 128. Sands BE, Sandborn WJ, Panaccione R, et al. Ustekinumab as in-
meta-analysis: anti-tumor necrosis factor α therapy and car- duction and maintenance therapy for ulcerative colitis. N Engl J
diovascular events in rheumatoid arthritis. Arthritis Care Res Med 2019;381:1201–14.
2011;63:522–9. 129. Szymanski CJ, Munusamy P, Mihai C, et al. Shifts in oxida-
113. Spanakis E, Sidiropoulos P, Papadakis J, et al. Modest but sus- tion states of cerium oxide nanoparticles detected inside intact
tained increase of serum high density lipoprotein cholesterol levels hydrated cells and organelles. Biomaterials 2015;62:147–54.
in patients with inflammatory arthritides treated with infliximab. 130. Krieckaert CLM, Nair SC, Nurmohamed MT, et al. Personalised
J Rheumatol 2006;33:2440–6. treatment using serum drug levels of adalimumab in patients
114. Chen DY, Chen YM, Hsieh TY, Hsieh CW, Lin CC, Lan JL. Sig- with rheumatoid arthritis: an evaluation of costs and effects. Ann
nificant effects of biologic therapy on lipid profiles and insulin Rheum Dis 2015;74:361–8.
resistance in patients with rheumatoid arthritis. Arthritis Res Ther 131. Eslam M, Newsome PN, Sarin SK, et al. A new definition for met-
2015;17:52. abolic dysfunction-associated fatty liver disease: an international
115. Ferraz Filho AC, dos Santos LP, Silva MB, Skare TL. Lipid profile expert consensus statement. J Hepatol 2020;73:202–9.
and anti-TNF-α use. Rev Bras Reumatol 2013;53:444–7. 132. Polyzos SA, Kang ES, Tsochatzis EA, et al. Commentary: nonalco-
116. Daïen CI, Duny Y, Barnetche T, Daurès JP, Combe B, Morel J. holic or metabolic dysfunction-associated fatty liver disease? The
Effect of TNF inhibitors on lipid profile in rheumatoid ar- epidemic of the 21st century in search of the most appropriate
thritis: a systematic review with meta-analysis. Ann Rheum Dis name. Metabolism 2020;113:154413.
2012;71:862–8. 133. Mintziori G, Polyzos SA. Emerging and future therapies for non-
117. Koutroubakis IE, Oustamanolakis P, Malliaraki N, et al. Effects alcoholic steatohepatitis in adults. Expert Opin Pharmacother
of tumor necrosis factor alpha inhibition with infliximab on lipid 2016;17:1937–46.

You might also like