Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2019-318536 on 29 November 2019. Downloaded from http://gut.bmj.com/ on November 30, 2019 at Lund University Libraries. Protected by copyright.
Novel concepts in the pathophysiology and treatment
of functional dyspepsia
Lucas Wauters,1,2 Nicholas J Talley  ‍ ‍,3,4 Marjorie M Walker,5 Jan Tack,1,2
Tim Vanuytsel1,2

1
Gastroenterology and ABSTRACT
Hepatology, University Hospitals Emerging data increasingly point towards the duodenum Key messages
Leuven, Leuven, Belgium
2
Translational Research in as a key region underlying the pathophysiology of
functional dyspepsia (FD), one of the most prevalent ►► Functional dyspepsia (FD) refers to a common
Gastrointestinal Disorders, KU
Leuven, Leuven, Belgium functional GI disorders. The duodenum plays a major unexplained disorder characterised by
3
Faculty of Health and Medicine, role in the control and coordination of gastroduodenal epigastric pain and/or postprandial discomfort
University of Newcastle, (fullness, early satiety).
function. Impaired duodenal mucosal integrity and low-­
Newcastle, New South Wales, ►► The major FD subgroups are the postprandial
Australia grade inflammation have been associated with altered
4
School of medicine and neuronal signalling and systemic immune activation, distress syndrome and epigastric pain syndrome
public Health, Hunter Medical and these alterations may ultimately lead to dyspeptic (PDS and EPS, respectively). PDS is the most
Research Institute, New symptoms. Likely luminal candidates inducing the prevalent subgroup but often overlaps with
Lambton Heights, New South EPS in those who consult and frequently co-­
Wales, Australia duodenal barrier defect include acid, bile, the microbiota
5
Anatomical Pathology, and food antigens although no causal association exists with gastro-­oesophageal reflux and IBS,
University of Newcastle, with symptoms has been convincingly demonstrated. resulting in complex and under-­recognised
Newcastle, New South Wales, Recognition of duodenal pathology in FD will hopefully symptom patterns.
Australia ►► Duodenal mucosal hyperpermeability and
lead to the discovery of new biomarkers and therapeutic
targets, allowing biologically targeted rather than low-­grade duodenal inflammation have been
Correspondence to associated with altered neuronal signalling and
Professor Nicholas J Talley, symptom-­based therapy. In this review, we summarise
Health, University of Newcastle, the recent advances in the diagnosis and treatment of FD systemic immune activation in FD.
Callaghan, NSW 2305, with a focus on the duodenum. ►► Psychiatric comorbidity such as anxiety and
Australia; depression is common and may arise from gut
​Nicholas.​Talley@n​ ewcastle.​ dysfunction or immune activation, or in other
edu.a​ u cases alter gastroduodenal function potentially
Received 29 May 2019
Introduction leading to dyspeptic symptom generation.
Revised 18 November 2019 Functional dyspepsia (FD) is a chronic GI disorder ►► Recognition of subtle pathology in FD may lead
Accepted 19 November 2019 defined by upper abdominal symptoms consid- to the discovery of new potential biomarkers
ered to originate from the gastroduodenal region and therapeutic targets, ultimately contributing
with no structural disease on routine investigation, to diagnosis and treatment.
including upper GI endoscopy.1 Two subgroups ►► Future classifications should include markers of
of FD were proposed by the Rome III consensus underlying pathophysiology to allow targeted
and following publication of additional supporting rather than symptom-­based therapy and to
evidence, reiterated in the Rome IV version: post- identify subgroups that respond to specific
prandial distress syndrome (PDS) with postpran- interventions.
dial fullness or early satiation, and epigastric pain
syndrome (EPS) with epigastric pain or epigastric
burning.2 In addition, symptoms must be severe overlap).3 Whereas PDS is characterised by meal-­
enough to impact on usual activities with a minimal related symptoms, epigastric pain and/or burning in
frequency of 1 (EPS) or 3 (PDS) days per week and EPS may be unrelated to the meal although patients
present for 3 months with symptom onset at least often under-­ report meal-­
related pain because it
6 months before diagnosis (table 1).2 Patients with may be delayed after eating.5 Although the PDS-­
FD have a reduced quality of life and experience EPS overlap group is still substantial, recognising
increased healthcare costs with loss of productivity, postprandial symptoms (including epigastric pain
confirmed by a recent cross-­sectional population-­ or burning) as part of the PDS subgroup in Rome
based study.3 IV substantially reduced overlap compared with the
© Author(s) (or their A meta-­ analysis using a broad definition Rome III definition.2 6 In FD, symptoms of upper
employer(s)) 2019. No reported that the global prevalence of uninvesti- abdominal bloating, belching, heartburn and nausea
commercial re-­use. See rights
and permissions. Published gated dyspepsia is 21%, with a higher prevalence can be present, although functional nausea and
by BMJ. in women, smokers, users of non-­ steroidal anti-­ vomiting, and functional bloating are categorised
inflammatory drugs (NSAIDs) and in those with separately.
To cite: Wauters L, Talley NJ,
Helicobacter pylori (Hp) infection.4 Recent figures Symptoms of heartburn and/or IBS often co-­exist
Walker MM, et al. Gut Epub
ahead of print: [please from the USA, Canada and UK showed a 10% prev- with FD. Heartburn in FD may be due to patholog-
include Day Month Year]. alence of FD in the adult population using the Rome ical acid reflux or functional heartburn.7 Although
doi:10.1136/ IV criteria, with a similar distribution pattern across functional heartburn (12%) and IBS (32%)
gutjnl-2019-318536 different regions (61% PDS, 18% EPS and 21% were independent factors associated with all FD
Wauters L, et al. Gut 2019;0:1–10. doi:10.1136/gutjnl-2019-318536    1
Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2019-318536 on 29 November 2019. Downloaded from http://gut.bmj.com/ on November 30, 2019 at Lund University Libraries. Protected by copyright.
peripheral manifestations from an Australian longitudinal study
Box 1  FD subgroups and overlap illustrated that depression without GI symptoms at baseline
predicted FD, whereas anxiety and depression developed in FD
►► Distribution of functional dyspepsia (FD) subtypes was similar patients without psychological comorbidity at baseline after 12
across countries (61% postprandial distress syndrome (PDS), years of follow-­up.19 Similar results were obtained in an inde-
18% epigastric pain syndrome and 21% overlap) with a 10% pendent population sample.20 These findings indicate an oppor-
prevalence in the adult population (Rome IV). tunity for prevention, as an average period of more than 3 years
►► PDS is characterised by meal-­related symptoms and was noted before the development of GI symptoms in patients
recognising postprandial symptoms (eg, epigastric pain or with mood or anxiety disorders.21
burning) as part of the PDS subgroup reduces overlap.
►► Epigastric bloating, belching and nausea as well as weight Gastric sensorimotor abnormalities
loss are part of the dyspepsia spectrum, but vomiting Abnormalities of gastric function including delayed emptying,
suggests an alternative diagnosis. impaired accommodation and hypersensitivity to distention have
►► Symptoms of heartburn and IBS are independently associated been reported in FD, but these changes correlate poorly or not at
with all FD subtypes and especially the overlap group, which all with symptoms.22 Moreover, the prevalence of altered gastric
may be related to duodenal eosinophilia. sensorimotor function was similar in the Rome III-­defined PDS

Table 1  Rome IV criteria for FD


Rome IV criteria Frequency, duration and onset Remarks and other symptoms
FD Bothersome postprandial fullness, early ≥1 symptom for the past 3 months with Vomiting suggests another disorder. Symptoms should not be
satiation, epigastric pain or burning AND onset ≥6 months before diagnosis relieved by evacuation of faeces or gas. Symptoms of GORD and
no evidence of structural disease likely to IBS may coexist with FD (PDS and EPS).
explain symptoms
PDS Bothersome postprandial fullness and/or ≥3 days/week for the past 3 months with Postprandial epigastric pain or burning, epigastric bloating,
early satiation severe enough to interfere onset ≥6 months before diagnosis excessive belching, nausea and heartburn can be present.
with daily activities or to prevent finishing
a meal
EPS Bothersome epigastric pain and/or epigastric ≥1 day/week for the past 3 months with Postprandial epigastric bloating, belching and nausea can be
burning severe enough to interfere with daily onset ≥6 months before diagnosis present. Pain may be induced or relieved by ingestion of a meal or
activities occur while fasting and does not fulfil biliary pain criteria.
EPS, epigastric pain syndrome; FD, functional dyspepsia; PDS, postprandial distress syndrome.

subtypes, the strongest association was found with the overlap-


ping group.3 In the Swedish Kalixanda study, overlap of FD was and EPS subgroups in a large tertiary care study, suggesting a
present with GORD in 4%, IBS in 2% and both in 6%.8 In a limited contribution to symptom generation.22 Although cardinal
follow-­up study, new-­onset GORD was common in FD with a FD symptoms did not correlate with gastric emptying,23 24 a
higher risk in patients with duodenal eosinophilia.9 10 Moreover, recent systematic review and meta-­analysis supported associa-
a population-­based study from Mayo Clinic concluded that FD is tions between upper GI symptoms and gastric emptying when
often underdiagnosed and mislabelled as GORD (box 1).11

Translational science concepts


Brain-gut interactions
Functional GI disorders are classified by the Rome IV criteria
as disorders of gut-­brain interaction with contributions of both
altered brain processing and luminal changes including dysbi-
osis (figure 1).12 13 Up to half of the subjects with uninvestigated
dyspepsia in the general population identify stress as a trigger for
their symptoms.14 There is evidence to support that the effect of
stress may be mediated by increased permeability and immune
activation.15 Recent preclinical studies indicate that stress can
lead to intestinal dysbiosis, which in turn affects central nervous
system function and behaviour,16 and these findings have led
to the concept of the bidirectional ‘brain-­ gut axis’ although
supporting human data are scarce. A systematic review of func-
tional neuroimaging studies in FD concluded that several brain
regions including the frontal and somatosensory cortex showed
anomalies.17 Abnormal central modulation (brain to gut) and
overactive visceral sensory signalling (gut to brain) may both
be involved in the pathophysiology of FD, and these pathways
are likely activated or modulated through psychological factors Figure 1  The gut-­brain axis in FD and mechanisms of overlap
and the hypothalamic-­pituitary-­adrenal axis stress response.17 18 with GORD and IBS. FD, functional dyspepsia; TLESR, transient lower
Evidence for a bidirectional interaction between central and oesophageal sphincter relaxation.
2 Wauters L, et al. Gut 2019;0:1–10. doi:10.1136/gutjnl-2019-318536
Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2019-318536 on 29 November 2019. Downloaded from http://gut.bmj.com/ on November 30, 2019 at Lund University Libraries. Protected by copyright.
optimal test methods were used.25 The distinction with gastropa- hypothesised. Therefore, gastric disturbances could be secondary
resis should be made as nausea may be present but vomiting is and not primary in FD.35 Also, altered duodeno-­gastric reflexes
unusual in FD,2 and nausea and vomiting are significant symp- and impaired gastric accommodation may underlie co-­existing
toms in idiopathic and diabetic gastroparesis.26 Gastric hyper- reflux symptoms through increased transient lower oesophageal
sensitivity and impaired accommodation may overlap, and no sphincter relaxations leading to new-­onset GORD, as heartburn
association with symptoms was found for (dys-­)accommodation was found to develop more often in patients with duodenal
except for severe PDS symptoms.22 27 Indeed, dyspeptic symp- pathology including duodenal eosinophilia.10 36 Moreover, it has
toms were significantly associated with psychosocial factors such also been suggested that overlapping IBS symptoms in FD could
as depression and somatisation and to a lesser extent with gastric be explained by more extensive intestinal inflammation with
sensorimotor function.28 As impaired gastric accommodation alteration of neuronal signalling and visceral hypersensitivity37
was reported in 43% of 1267 patients with functional gastro- (figure 1).
duodenal symptoms seen at a single tertiary centre and half of
these patients also had abnormal gastric emptying, the study and
treatment of gastric motor dysfunction likely remain relevant in Duodenal barrier defect and immune activation
at least a subset of patients.29 The duodenum has emerged as a key player in both GI and
Emerging data increasingly point towards the duodenum as metabolic diseases as it regulates the passage of food as chyme
the key integrator in dyspepsia symptom generation, and it has from the stomach to the small intestine, where nutrients
been proposed that gastric motor dysfunction may be attributed are absorbed.38 Autocrine and paracrine mechanisms in the
to disordered duodeno-­ gastric feedback.30 31 Several studies duodenum are also involved in the mucosal defence to acid
have now reported duodenal mucosal low-­grade inflammation and the luminal digestion of nutrients with secretion of bile
and increased small intestinal homing T cells as markers of and pancreatic juice.39 Activation of duodeno-­ gastric feed-
intestinal inflammation, which correlated with delayed gastric back mechanisms by chemical or mechanical triggers influ-
emptying and dyspeptic symptom severity.32 33 Evidence for ences gastric emptying, with an important role of intestinal or
the role of duodenal inflammation in neuronal signalling in FD pancreatic hormones including incretins and gastric orexigenic
comes from a study in Leuven showing correlations between hormones, which also signal to the brain.40 Besides nutrient
duodenal eosinophils or mast cells and calcium transient ampli- sensing, transmucosal passage of luminal content is likely in
tudes to high K+ or electrical stimulation.34 The exact mecha- the proximal small intestine due to regional variation in the
nism through which duodenal inflammation induces neuronal mucosal barrier along the GI tract and the crypt-­villus axis,
hyperexcitability is still incompletely understood, but in analogy ranging from >20 angstrom at the crypt base to 5 angstrom
to IBS, a role for histamine and proteases—among others—is at the villus tip in the proximal small intestine, which is also

Figure 2  The duodenal microenvironment in health and FD with pathophysiological mechanisms. In health (left), duodenal luminal content is
separated from the mucosal immune system by an intact duodenal barrier. In case of duodenal barrier dysfunction such as observed in FD (right),
luminal triggers from altered duodenal content and/or dysbiosis may lead to both local and systemic immune activation with disturbed signalling
of duodeno-­gastric feedback mechanisms by neuronal changes in afferent nerves. These changes in the duodenal microenvironment may generate
dyspeptic symptoms. Arrows (red) indicate impaired duodenal mucosal integrity and eosinophil-­mast cell signalling. FD, functional dyspepsia.
Wauters L, et al. Gut 2019;0:1–10. doi:10.1136/gutjnl-2019-318536 3
Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2019-318536 on 29 November 2019. Downloaded from http://gut.bmj.com/ on November 30, 2019 at Lund University Libraries. Protected by copyright.
the most permeable region with the largest intercellular pores structural submucosal neuronal changes have indeed been
of the GI tract.41 Although the small bowel microenvironment reported in the duodenum of FD patients, correlating with
with a loose glycocalyx (which covers the brush border on the accumulation of eosinophils and mast cells in close proximity
apical surface of the epithelial cells) allows for a closer inter- to neurons.34 Activation and degranulation of eosinophils in
action between the lumen and the host cells in comparison the presence of normal numbers may also result in neuronal
with the colon, the defence against potential noxious luminal changes with dyspeptic symptom generation.44 58 59 Although
factors is reflected in the barrier function and distribution systemic markers of inflammation were elevated and linked
of immune cells42 (figure 2). Ussing chamber experiments with both duodenal hyperpermeability and gastric emptying as
have shown an increased duodenal mucosal permeability well as dyspeptic symptoms,33 57 local hypersensitivity in the
in FD with a lower transepithelial electrical resistance and duodenum could also be implicated in FD.60 61
higher paracellular passage of fluorescent-­ labelled dextran The most attractive hypothesis for symptom generation is
molecules.32 The expression of particular cell-­ to-­
cell adhe- loss of mucosal integrity as a primary event, which could lead
sion proteins, including tight junctions (ZO-1 and occludin), to immune activation through antigen presentation with a
adherins junctions (β-catenin and E-­ cadherin) and desmo- predominant T-­helper type 2 (Th2) or a Th17 response leading
somes (desmoglein-2), was also significantly decreased and to recruitment and degranulation of eosinophils, triggering
correlated with duodenal hyperpermeability and the severity visceral hypersensitivity and altered motor control1 (figure 2).
of low-­grade mucosal inflammation, including duodenal mast Eosinophils are critical effector cells of a Th2 response charac-
cells and eosinophils.32 This study of FD patients from Leuven terised by Th2 allergic inflammation.62 The consistent finding
(Belgium) confirmed the initial reports on duodenal eosin- of duodenal eosinophilia may help explain the observed link
ophilia in adults from Sweden43 and activation of duodenal with atopy in a UK-­based primary care registry of functional
eosinophils in children in an uncontrolled pilot study from the GI disorders including FD63 and also autoimmune disease.64
USA.44 Studies using duodenal mucosal impedance measure- The association with allergy may indicate a hypersensitivity
ment also showed lower values in FD patients although the reaction in some patients with early satiety.65 Also, associa-
comparison with controls did not reach statistical significance tions of dyspeptic symptoms in the general population with
after adjusting for potential confounders (eg, NSAIDs).45 herbivore but not carnivore pets and antibiotics suggest the
Mucosal impedance values were also positively correlated involvement of microbiota-­ related components in the gut
with ZO-1 (tight junction protein) and inversely correlated although this was not tested and further studies should control
with tissue interleukin (IL)-­1b expression, implicating the role for allergy and other atopic conditions.66 Thus, a luminal
of low-­grade inflammation in duodenal barrier dysfunction.45 trigger (food, microbiota or secreted endogenous mediators)
However, challenges remain in the interpretation and repro- in the duodenum may cause the observed mucosal changes in
ducibility of the experimental methods. One of the limitations FD patients.
of the ex vivo studies is that only the integrity of the epithelial
layer is measured, lacking both the superficial mucus layer and
innervation by the enteric nervous system, which also affect Duodenal acid, duodenal bile and dysbiosis
mucosal barrier function. Although the causes of the barrier defect and immune activation
The local and systemic inflammatory changes in FD were in FD are still unknown, likely candidates include duodenal acid,
recently summarised in two systematic reviews46 47 (table 2). bile, stress and food or microbial components. Duodenal acid
Mast cells have been implicated in the pathophysiology of perfusion resulted in delayed gastric emptying, impaired accom-
IBS and stress-­ induced small intestinal hyperpermeability, modation and hypersensitivity to distension in healthy subjects,
measured with the urinary lactulose-­mannitol ratio in healthy suggesting a potential role for duodenal acid exposure and alter-
students undergoing psychological stress during an oral exam- ations in gastric sensorimotor function although these perfusion
ination.15 The finding of duodenal mast cells in the Leuven experiments do not reflect physiological changes occurring in
cohort may be explained by overlapping FD and IBS, as mast the distal duodenum.67–69 Although gastric acid secretion in FD
cells in the duodenum were characteristic of IBS in another is normal, an increased duodenal acid exposure, possibly due to
study.48 The finding of duodenal eosinophilia49–51 was accom- delayed duodenal acid clearance, has been reported with acid
panied by cytotoxic T cell52 or mast cell32 34 50 53 infiltration in perfusion experiments in FD patients.60 70 Moreover, duodenal
a subset of studies. Eosinophil-­mast cell signalling may also be acid perfusion in healthy subjects resulted in both gastric relax-
involved in stress-­induced hyperpermeability as administration ation and duodenal mucosal hyperpermeability and mast cell
of corticotropin-­release hormone, which can activate specific activation, which were however not reversed by pretreatment
receptors on both eosinophils and mast cells,54 increased with DSCG.71 The role of food in triggering FD symptoms and its
small intestinal permeability, and this effect was blocked by role in duodenal low-­grade inflammatory changes have remained
pretreatment with the mast cell stabiliser disodium cromogly- unexplored to date. Duodenal hypersensitivity to lipids has been
cate (DSCG).15 Increased duodenal mucosal eosinophil counts reported in FD,30 with modulation of upper GI symptoms via
have been associated with postprandial symptoms in Austra- cholecystokinin signalling in response to fat.61 The role of gluten
lian adults55 and children.56 The relation between duodenal in FD has not been investigated although symptoms may overlap
eosinophilia and meal-­ related symptoms may be explained with non-­celiac gluten sensitivity, in which duodenal eosinophils
by altered duodeno-­ gastric reflexes and gastric dysaccom- may also cause gastric sensorimotor dysfunction (box 2).72
modation in response to the meal, although studies directly The release of bile salts in the duodenum has been impli-
correlating duodenal abnormalities and gastric sensorimotor cated in the onset or worsening of dyspeptic symptoms after
dysfunction are scarce.48 55 Recently, duodenal hyperperme- a meal.73 Bile salts and bacteria have a bidirectional relation-
ability (measured in Ussing chambers) was correlated with ship since specific bile salts have antimicrobial effects against
gastric emptying to solids in an FD patient cohort from Leuven selected bacteria and bacteria are responsible for bile salt trans-
(Belgium), supporting the hypothesis of altered duodeno-­ formation, mainly in the colon with reabsorption and reconju-
gastric feedback in symptom generation.57 Functional and gation of primary and secondary bile salts before excretion in
4 Wauters L, et al. Gut 2019;0:1–10. doi:10.1136/gutjnl-2019-318536
Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2019-318536 on 29 November 2019. Downloaded from http://gut.bmj.com/ on November 30, 2019 at Lund University Libraries. Protected by copyright.
but conclusive evidence is lacking and more in-­depth studies are
Box 2 New observations in the duodenum needed to elucidate the pathophysiological mechanism.
Immune activation in the pathogenesis of FD is very evident in
►► Duodenal changes including mucosal hyperpermeability and the postinfectious setting,77 with persisting changes in duodenal
low-­grade inflammation have been associated with altered mucosal immune cells after the initial event and systemic immune
neuronal signalling and systemic immune activation. activation in acute compared with unspecified-­onset FD, indicating
►► Immune activation has been observed, and patients
the inability of the immune system to recover from the triggering
with functional dyspepsia may have an allergic or atopic
infectious insult.77 78 Similar to studies in IBS, antibodies to cyto-
background with a Th2-­predominant immune profile.
lethal distending toxin B, produced by Gram-­negative bacteria
►► The cause of the barrier dysfunction is unknown and
causing acute gastroenteritis, were more common in FD and
likely candidates include duodenal acid, bile salts, food
IBS/FD overlap compared with healthy controls in an Australian
components, stress and the mucosal microbiome.
►► Gastric dysfunction may be driven by impaired duodeno-­
population-­based study, suggesting possible under-­recognition of
gastric feedback mechanisms through local signalling or postinfectious FD.79 The disruption in structural and/or func-
changes in systemic immune activation. tional microbial configuration, defined as ‘dysbiosis’, has been
studied in both functional and inflammatory GI disorders.80 81
Culture-­ independent metagenomics and high-­ throughput 16S
the duodenum.74 Although bacterial deconjungation may also ribosomal RNA gene sequencing have revolutionised our under-
occur in the small bowel, deconjugated bile salts were only standing of the human gut microbiome or collective genome of
reported at or below the detection limit in nasoduodenal aspi- microorganisms inhabiting the GI tract.82 However, the focus
rates from FD patients with decreased fasted concentrations of has almost been exclusively on faecal microbiota, which may not
primary bile salts.75 Moreover, a decreased ratio of primary to reflect the mucosa-­associated microbiota (MAM).83 In addition,
secondary bile salts was found during fed state in FD patients the duodenal microbiome has hardly been studied although the
compared with controls, correlating with duodenal mucosal commensal gut microbiota of the small intestine play an essen-
permeability although secondary bile salt concentrations were tial role in nutrient acquisition, colonisation resistance to patho-
similar between groups.76 These data suggest that the relative gens, immune development and epithelial barrier function.42
abundance of primary versus secondary bile salts, rather than The combination of gastric acid, bile, digestive enzymes and
their absolute concentrations, affects intestinal barrier function rapid transit time in the duodenum may contribute to a hostile

Table 2  Mucosal and systemic immune activation in FD


Findings FD subtype Reference
Mucosal immune cells Duodenal eosinophil infiltration NUD (FD) Talley et al 200743
PDS Walker et al 201065
Walker et al 201455
Paediatric Friesen et al 200244
Wauters et al 201744
PI-­FD Futagami et al 201049
Unspecified Walker et al 200948
Vanheel et al 201432
Cirillo et al 201534
Wang et al 201550
Du et al 201658
Tanaka et al 201651
Duodenal eosinophil degranulation Paediatric Friesen et al 200244
Unspecified Du et al 201658
Vanheel et al 201859
Duodenal mast cell infiltration FD-­IBS overlap Walker et al 200948
Unspecified Vanheel et al 201432
Cirillo et al 201534
Wang et al 201550
Yuan et al 201553
Duodenal mast cell degranulation Unspecified Vanheel et al 201859
Duodenal (CCR2+) macrophage infiltration PI-­FD Futagami et al 201049
Kindt et al 200977
Duodenal CD8+T-­cell infiltration Unspecified Gargala et al 200752
Decrease in CD4+ cells PI-­FD Kindt et al 200977
No change in CD3+ lymphocyte
Mucosal cytokines No studies
Circulating immune cells CD45RA+ (naive)/ CD45RO+ (activated) lymphocytes PI-­FD Kindt et al 200978
α4β7-­T cells (gut-­homing) Unspecified Liebregts et al 201133
Circulating cytokines IL-10 PI-­FD Kindt et al 200978
IL-1β and TNF-­a Unspecified Liebregts et al 201133
FD, functional dyspepsia;IL, interleukin ; NUD, non-­ulcer dyspepsia; PDS, postprandial distress syndrome; PI-­FD, post-­infectious functional dyspepsia; TNF, tumour necrosis factor.

Wauters L, et al. Gut 2019;0:1–10. doi:10.1136/gutjnl-2019-318536 5


Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2019-318536 on 29 November 2019. Downloaded from http://gut.bmj.com/ on November 30, 2019 at Lund University Libraries. Protected by copyright.
Figure 3  Old and new treatments based on potential pathways and therapeutic targets in functional dyspepsia. Effective treatments are indicated
with green boxes and arrows, with solid lines for evidence from randomised controlled trials and dashed lines for evidence from non-­randomised
controlled trials. Red boxes indicate potentially harmful side-­effects of treatments. PPI, proton pump inhibitor.

environment with lower density but greater diversity in the studies45) and neuronal function (eg, calcium imaging34), new
duodenal bacterial flora compared with the rectum.84 Although biomarkers from duodenal biopsies may include mucosal tight
more challenging to study because of the lower bacterial abun- junction proteins32 or IL-­1B expression.45 Nevertheless, none of
dance and difficult sampling, host-­ microbiome interactions these advanced tests can be recommended for routine clinical
could be more important in the small intestine with predom- practice since their outcome does not affect the management.
inant Gram-­positive aerobes in the duodenum compared with The routine use of gastric function testing is questionable as
obligate anaerobes in the colon.80 Data on the duodenal MAM discussed below.
in FD are currently limited to one pilot study involving nine The complexity of the syndrome indicates a multifactorial
FD patients, with an increase in Streptococcus and total bacte- origin, and the lack of effective therapies has been recognised
rial load in the duodenal MAM compared with controls, which by international societies such as the American College of
correlated with meal-­related symptom severity and quality of Gastroenterology (ACG) and Canadian Association of Gastro-
life.85 However, sample size was small with no information on enterology (CAG), which have recently issued guidelines.90 As
medication, which is an important confounder, nor IBS overlap, discussed below, the commonly used therapies affect brain-­gut
which has also been associated with faecal and duodenal dysbi- or gastric pathways, which do however not always correlate with
osis.80 84 86 87 Interestingly, the abundance of Veillonella in the symptoms and only limited treatments target the underlying
duodenal MAM was negatively correlated with gastric emptying duodenal pathophysiology (figure 3). In addition, an increased
time,88 but replication and further studies are needed in order to awareness of side effects has limited the routine use of some
unravel the potential role of the gut microbiota in FD. drugs, including neuromodulators and prokinetics. Evidence for
dysbiosis and overgrowth with oral species with proton pump
Advances in clinical practice inhibitors (PPI)91 92 has provided evidence for changes in the gut
Investigation and management environment although the link with GI symptoms is still unclear.
A systematic review and meta-­ analysis reported that endo- Moreover, although still early days, randomised controlled trials
scopic investigation for dyspeptic symptoms was normal in over of probiotics and selective antibiotics have shown improvement
70% of referrals, thus leading to a diagnosis of FD.89 Although in both symptoms and the microbiome of FD patients.93–95
peptic ulcers and gastric cancer have become uncommon and
rarer causes of dyspepsia, upper endoscopy with duodenal and Targeting brain-gut and gastric abnormalities
gastric biopsies is still the preferred diagnostic method for the The efficacy of neuromodulators versus placebo (risk ratio (RR)
exclusion of coeliac disease and Hp infection, which may also 0.78; 95% CI 0.68 to 0.91; number needed to treat (NNT)=6)
cause dyspeptic symptoms.1 Following Rome IV criteria, the was limited to antipsychotics and tricyclic antidepressants
diagnosis of FD can only be made in the absence of structural (TCAs) in a recent systematic review and meta-­analysis of 13
disease including upper endoscopy,2 but this does not exclude studies including 1241 patients.96 In the Functional Dyspepsia
the presence of microscopic changes such as duodenal eosino- Treatment Trial, amitriptyline but not escitalopram was effec-
philia. Based on a control group selected from 1001 community tive for the treatment of ulcer-­like (painful) FD, with a three-
subjects in Sweden,43 a cut-­off of 22 eosinophils per 5 high-­power fold increased odds of reporting adequate relief of symptoms
fields was proposed for adults,65 although results can also be compared with placebo.97 In addition, FD patients with delayed
expressed per mm2 as shown in adult patients,55 and a duodenal gastric emptying did not respond to amitriptyline and this was
eosinophil count of >112 per mm2 was associated with a 33-­fold not related to a treatment-­induced delay in gastric emptying,
increased risk of FD in a paediatric Australian cohort.56 Due to indicating an effect on visceral sensitivity and abdominal pain
the limited availability of experimental investigations for testing rather than gastric motility.97 98 However, little is known on
duodenal barrier (eg, Ussing chambers32 or mucosal impedance the longer-­term efficacy of neuromodulators as the longest
6 Wauters L, et al. Gut 2019;0:1–10. doi:10.1136/gutjnl-2019-318536
Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2019-318536 on 29 November 2019. Downloaded from http://gut.bmj.com/ on November 30, 2019 at Lund University Libraries. Protected by copyright.
treatment duration was 12 weeks.96 The tetracyclic antidepres- addition, anti-­inflammatory effects such as the downregulation
sant mirtazapine significantly improved PDS symptoms, quality of eotaxin gene transcription seen in eosinophilic oesophagitis
of life, nutrient tolerance and body weight in a randomised trial may also reduce duodenal eosinophilia as recently shown in a
of 34 FD patients with significant weight loss (>10% of orig- cross-­sectional study,107 although this requires confirmation in a
inal body weight) and no depression or anxiety, which could prospective setting, using the Rome IV-­defined subgroups.
be linked to the antagonism of histamine-1, adrenergic α2 and Acid suppression with histamine-2 receptor antagonists
both serotonin-­2C and serotonin-3 receptors.99 Finally, treat- (H2RA) is also possible with a lower NNT of 7 compared with
ment with buspirone, a serotonin-­1A receptor agonist, which placebo-­ controlled studies of PPI according to a Cochrane
relaxes the proximal stomach in healthy individuals, signifi- review from 2006 with inclusion of older studies.108 In contrast,
cantly reduced both overall and PDS-­type symptoms in FD and the recent Cochrane meta-­analysis also included studies directly
improved gastric accommodation.100 Although the solid gastric comparing PPI and H2RA with no difference between both
emptying rate was unchanged, a delay in liquid gastric emptying treatments (RR 0.88; 95% CI 0.74 to 1.04).105 However, due to
was noted but the relevance for dyspeptic symptoms is probably the lack of high-­quality comparative trials and superior antise-
limited.100 cretory effect of PPIs, the ACG/CAG guidelines suggest PPI over
Prokinetics enhance gastric emptying, and although delayed H2RA as first-­line therapy,90 although both are proposed in the
gastric emptying and impaired gastric accommodation may be NICE guidelines.103 Anti-­inflammatory effects of H1RA due to
more common in PDS patients, this was not confirmed in a blockade of histamine have been studied in the generation of
large study that showed a similar prevalence of gastric motor visceral hypersensitivity in IBS109 and may also prove benefi-
abnormalities in Rome III-­defined FD subgroups.22 Moreover, cial in FD patients with duodenal mast cell infiltration. More-
a systematic analysis of 34 studies showed no evidence of a over, combined histamine-1 and histamine-2 receptor blockade
correlation between the acceleration of gastric emptying and with the mast cell stabiliser cromoglycate in non-­responders led
symptom improvement in the treatment of diabetic and idio- to an overall response rate of 90% in dyspeptic children with
pathic gastroparesis.101 Delayed gastric emptying may be present duodenal eosinophilia who previously had failed on H2RA.110
in about 23% of FD patients, and prokinetics also affect gastric The beneficial effect of combined histamine-1 and histamine-2
accommodation and sensitivity, which were disturbed in 37% of receptor blockade was also reported in a recent retrospective
FD patients.22 These effects may explain the significant benefit case series from Australia with a trend for higher duodenal eosin-
of prokinetics in reducing ongoing dyspeptic symptoms (RR ophil counts at baseline in responders versus non-­responders.111
0.81; 95% CI 0.74 to 0.89; NNT=7) in a recent meta-­analysis Treatment with the leucotriene-1 receptor antagonist montelu-
of 29 studies involving 10 044 FD patients.102 However, signif- kast resulted in a 62% clinical response rate compared with 32%
icant heterogeneity and publication bias were noted and the on placebo (p<0.02). In another paediatric cohort,112 the short-­
overall effect was less convincing after removing cisapride from term clinical response of montelukast was not associated with
the meta-­analysis (NNT=12), which was withdrawn from the changes in eosinophil density or activation.113
market due to cardiac adverse events.102 Other prokinetics such
as domperidone and acotiamide are not widely available, and Targeting duodenal dysbiosis
extrapyramidal side effects (for dopamine receptor antagonists) Although the ACG/CAG and NICE guidelines advice against
and QTc prolongation with domperidone limit their chronic use, dose escalation in case of insufficient improvement with a one
leading the ACG/CAG guidelines to recommend TCA in PPI-­ time per day dose of PPI during 4–8 weeks, inappropriate and
refractory FD patients before prokinetics.90 prolonged use of PPIs even in the absence of clinical benefit is
frequently reported. Moreover, PPIs have been associated with
an increased risk of enteric infections (including Clostridium
Acid-suppressive and anti-inflammatory therapies difficile)114 and overabundance of oral or potentially patho-
Acid-­suppressive therapy with PPIs is recommended as first-­ genic flora in the gastric115 and faecal microbiome.91 Data
line treatment for FD by the ACG/CAG guidelines90 and the on the duodenal microbiota are still lacking. The increase in
National Institute for Health and Care Excellence (NICE)103 oral or potentially pathogenic flora on PPI was represented
after eradication of Hp or in Hp-­negative patients. Empirical by Streptococcus in the gastric MAM, and this may have
PPI therapy before a test-­and-­treat approach for Hp has also contributed to persisting dyspeptic symptoms in a subgroup of
been proposed in cases of low (<20%) Hp prevalence.104 In a
recent Cochrane meta-­analysis including 6172 patients, a reduc-
tion of global dyspeptic symptoms (RR 0.88; 95% CI 0.82 to
Box 3  Targeted treatments in FD
0.94; NNT=11) with similar quality of life on PPI compared
with placebo was reported.105 This was confirmed by the
►► Treatments for brain-­gut and gastric dysfunction, including
meta-­analysis of the ACG/CAG90 (NNT=10) with no differ-
neuromodulators and prokinetics, have a limited efficacy and
ences for different doses or types of PPI.90 105 According to
are hampered by side effects.
the Rome IV consensus, PPIs are considered ineffective for the
►► Proton pump inhibitors may have both acid-­suppressive
PDS subgroup,2 which was based on the results from an older
and anti-­inflammatory effects in functional dyspepsia (FD)
meta-­analysis including showing efficacy of PPIs in the epigas-
patients with duodenal eosinophilia and impaired barrier
tric pain but not the dysmotility-­like FD subgroups according
function.
to Rome I and II criteria.106 However, a tendency for a higher
►► Targeting duodenal inflammation and the eosinophil-­mast
efficacy of PPI in PDS (RR 0.89; 95% CI 0.77 to 1.03) versus
cell axis with mast cell stabilisers and antihistamines requires
EPS (RR 0.99; 95% CI 0.76 to 1.28) was shown in the recent
studies in adult populations.
Cochrane meta-­analysis although the number of included ‘pure
►► Duodenal dysbiosis in FD may be treated with selective
EPS’ patients was low.105 While the exact mechanism of action
antibiotics, such as rifaximin, but requires confirmation in
in PDS is unknown, PPIs are beneficial for overlapping GORD
Western populations.
symptoms and may also reduce duodenal acid exposure.30 In
Wauters L, et al. Gut 2019;0:1–10. doi:10.1136/gutjnl-2019-318536 7
Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2019-318536 on 29 November 2019. Downloaded from http://gut.bmj.com/ on November 30, 2019 at Lund University Libraries. Protected by copyright.
patients.116 Therefore, this study and others have questioned target symptoms and gastric sensorimotor function rather than
the use of PPI therapy in FD patients, as does their limited the underlying duodenal pathology.35 37
efficacy.90 These limited data suggest that stopping PPIs in Future classifications of FD subtypes should take into account
patients without a clear benefit may be a first option to target the different pathophysiological mechanisms in order to allow
dysbiosis in FD. Eradication therapy with antibiotics improves targeted and not just symptom-­based therapy. Recognition of
symptoms in Helicobacter pylori-­ related dyspepsia (HpD), immune activation will hopefully lead to the discovery of new
and the effect was greater with metronidazole and in patients biomarkers and therapeutic targets, ultimately contributing to
with microscopic duodenitis,117 118 suggesting an effect on the the diagnosis and treatment of this chronic and challenging GI
duodenal microbiome, which is distinct from Hp eradication condition. The effect of acid suppression on the microbiome
per se.119 is highly relevant as PPIs are frequently scrutinised because of
Probiotics may improve HpD by inhibition of Hp, but overprescribing and potential long-­term adverse events. PPI-­
their efficacy was also demonstrated in Hp-­ uninfected FD induced dysbiosis may also help explain the limited efficacy of
patients.93 94 Following a 12-­week period of daily yoghurt inges- long-­term PPIs as the current first-­line treatment in FD90 and
tion containing 109 colony-­forming units of Lactobacillus gasseri the possibility of persisting dyspeptic symptoms in a subgroup
OLL2716 (LG21) or placebo (fermented milk product without of patients on PPIs.117 Other treatments targeting the micro-
L. gasseri) in Hp-­negative FD patients, a trend for a positive biome in FD look promising but require confirmation.
overall effect on gastric symptoms (p=0.07) and significantly
higher elimination rates for the major FD symptoms including Contributors  LW drafted the manuscript. JT revised the manuscript. MMW revised
the manuscript. NJT drafted and revised the manuscript. TV drafted and revised the
PDS-­like but not EPS-­like symptoms was noted.93 In a follow-­up manuscript.
study, changes in the gastric fluid microbiota were reported in
Funding  LW and TV are supported by the Flanders Research Foundation (FWO
FD patients at baseline with a restoration after intake of LG21.94 Vlaanderen) through a doctoral fellowship and a senior clinical research mandate,
In addition, bile acids were more frequently detected in gastric respectively. JT is supported by a Methusalem grant of KU Leuven. NJT is supported
fluid samples from FD patients compared with controls and by an NHMRC Level 3 Investigator Grant and is lead investigator of an NHMRC
although bile acids did not decrease with intake of LG21, the Centre of Research Excellence.
presence of an intestinal-­like bacterial profile was restored after Competing interests  NT: Consultancy: Allergan, IM Health Sciences, Takeda,
probiotics.94 This study and others have suggested the presence Theravance, Danone, Sanofi. JT: Consultancy: AlfaWassermann, Allergan, Danone,
of small intestinal bacterial overgrowth (SIBO) in a subset of FD Shire, Takeda, Theravance, Tsumura and Zeria Pharmaceuticals; Speaker fees: Abbott,
Allergan, Shire, Takeda and Zeria Pharmaceuticals. TV: Speaker fees: Abbott. Research
patients,94 120 which could be a marker and target although no Grant: Danone.
standardised diagnosis is available.
Patient consent for publication  Not required.
Treatment with rifaximin, a non-­absorbable and selective anti-
biotic with established efficacy for the treatment of abdominal Provenance and peer review  Commissioned; externally peer reviewed.
pain and bloating in non-­constipated IBS,121 may reverse SIBO ORCID iD
as measured with hydrogen breath testing,122 but this putative Nicholas J Talley http://​orcid.​org/​0000-​0003-​2537-​3092
mechanism is highly controversial. In a recent randomised trial,
rifaximin was superior to placebo for the adequate relief of
both global dyspeptic symptoms (78% vs 52%) at 8 weeks and References
1 Talley NJ, Ford AC. Functional dyspepsia. N Engl J Med 2015;373:1853–63.
postprandial fullness, bloating and belching at 4 weeks in FD 2 Stanghellini V, Chan FKL, Hasler WL, et al. Gastroduodenal disorders.
patients who were Hp negative.95 The effect on symptoms was Gastroenterology 2016;150:1380–92.
more pronounced in women with a similar incidence of adverse 3 Aziz I, Palsson OS, Törnblom H, et al. Epidemiology, clinical characteristics, and
effects in the rifaximin and control group. Although the exact associations for symptom-­based Rome IV functional dyspepsia in adults in the USA,
mechanism of action is not known, the antibiotic effect and Canada, and the UK: a cross-­sectional population-­based study. Lancet Gastroenterol
Hepatol 2018;3:252–62.
increased solubility of rifaximin with bile salts could explain its 4 Ford AC, Marwaha A, Sood R, et al. Global prevalence of, and risk factors for,
therapeutic effect in FD patients with changes in the duodenal uninvestigated dyspepsia: a meta-­analysis. Gut 2015;64:1049–57.
microenvironment (box 3).123 5 Bisschops R, Karamanolis G, Arts J, et al. Relationship between symptoms and
ingestion of a meal in functional dyspepsia. Gut 2008;57:1495–503.
6 Carbone F, Holvoet L, Tack J. Rome III functional dyspepsia subdivision in PDS and
Conclusion Eps: recognizing postprandial symptoms reduces overlap. Neurogastroenterol Motil
2015;27:1069–74.
The paradigm of FD as the most common functional upper GI 7 Savarino E, Pohl D, Zentilin P, et al. Functional heartburn has more in common with
disorder with ‘no evidence of structural disease’ has recently functional dyspepsia than with non-­erosive reflux disease. Gut 2009;58:1185–91.
been challenged.32 33 43 56 78 Despite the common occurrence 8 Aro P, Talley NJ, Agréus L, et al. Functional dyspepsia impairs quality of life in the
of FD in up to 15% of the general population, the underlying adult population. Aliment Pharmacol Ther 2011;33:1215–24.
9 Aro P, Talley NJ, Johansson S-­E, et al. Anxiety is linked to new-­onset dyspepsia
pathophysiology remains unclear.1 2 30 Emerging data point
in the Swedish population: a 10-­year follow-­up study. Gastroenterology
towards the duodenum as the key integrator in symptom 2015;148:928–37.
generation. Meal-­related symptoms in the PDS and overlap 10 Ronkainen J, Aro P, Walker MM, et al. Duodenal eosinophilia is associated with
group are linked with increased duodenal mucosal permea- functional dyspepsia and new onset gastro-­oesophageal reflux disease. Aliment
bility and low-­grade inflammation with submucosal neuronal Pharmacol Ther 2019;50:24–32.
11 Pleyer C, Bittner H, Locke GR, et al. Overdiagnosis of gastro-­esophageal reflux
changes,32 34 43 55 providing a basis for postprandial dyspeptic
disease and underdiagnosis of functional dyspepsia in a USA community.
symptoms and a potential diagnostic or predictive tool. Neurogastroenterol Motil 2014;26:1163–71.
However, it is still unclear whether these changes are indeed 12 Drossman DA, Hasler WL. Rome IV—Functional Gi disorders: disorders of gut-­brain
causal or secondary to an unknown cause. The association interaction. Gastroenterology 2016;150:1257–61.
with GORD due to gastric disturbances secondary to duodenal 13 Tack J, Drossman DA. What’s new in Rome IV? Neurogastroenterol Motil
2017;29:e13053.
inflammation and the possibility of overlapping IBS depending 14 Piessevaux H, De Winter B, Louis E, et al. Dyspeptic symptoms in the general
on the extent of intestinal inflammation also require further population: a factor and cluster analysis of symptom groupings. Neurogastroenterol
study.10 Current treatment options are of limited efficacy and Motil 2009;21:378–88.

8 Wauters L, et al. Gut 2019;0:1–10. doi:10.1136/gutjnl-2019-318536


Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2019-318536 on 29 November 2019. Downloaded from http://gut.bmj.com/ on November 30, 2019 at Lund University Libraries. Protected by copyright.
15 Vanuytsel T, van Wanrooy S, Vanheel H, et al. Psychological stress and corticotropin-­ 45 Komori K, Ihara E, Minoda Y, et al. The altered mucosal barrier function in the
releasing hormone increase intestinal permeability in humans by a mast cell-­ duodenum plays a role in the pathogenesis of functional dyspepsia. Dig Dis Sci
dependent mechanism. Gut 2014;63:1293–9. 2019;64:3228–39.
16 De Palma G, Blennerhassett P, Lu J, et al. Microbiota and host determinants of 46 Du L, Chen B, Kim JJ, et al. Micro-­Inflammation in functional dyspepsia: a systematic
behavioural phenotype in maternally separated mice. Nat Commun 2015;6. review and meta-­analysis. Neurogastroenterol Motil 2018;30:e13304.
17 Lee I-­S, Wang H, Chae Y, et al. Functional neuroimaging studies in functional 47 B Biomed GB, Carroll G, Mathe A, et al. Evidence for local and systemic immune
dyspepsia patients: a systematic review. Neurogastroenterol Motil activation in functional dyspepsia and the irritable bowel syndrome: a systematic
2016;28:793–805. review. Am J Gastroenterol 2019;114:429–36.
18 Van Oudenhove L, Aziz Q. The role of psychosocial factors and psychiatric disorders 48 Walker MM, Talley NJ, Prabhakar M, et al. Duodenal mastocytosis, eosinophilia
in functional dyspepsia. Nat Rev Gastroenterol Hepatol 2013;10:158–67. and intraepithelial lymphocytosis as possible disease markers in the irritable bowel
19 Koloski NA, Jones M, Kalantar J, et al. The brain–gut pathway in functional syndrome and functional dyspepsia. Aliment Pharmacol Ther 2009;29:765–73.
gastrointestinal disorders is bidirectional: a 12-­year prospective population-­based 49 Futagami S, Shindo T, Kawagoe T, et al. Migration of eosinophils and CCR2-/CD68-­
study. Gut 2012;61:1284–90. double positive cells into the duodenal mucosa of patients with postinfectious
20 Koloski NA, Jones M, Talley NJ. Evidence that independent gut-­to-­brain and functional dyspepsia. Am J Gastroenterol 2010;105:1835–42.
brain-­to-­gut pathways operate in the irritable bowel syndrome and functional 50 Wang X, Li X, Ge W, et al. Quantitative evaluation of duodenal eosinophils and mast
dyspepsia: a 1-­year population-­based prospective study. Aliment Pharmacol Ther cells in adult patients with functional dyspepsia. Ann Diagn Pathol 2015;19:50–6.
2016;44:592–600. 51 Tanaka F, Tominaga K, Fujikawa Y, et al. Concentration of glial cell line-­derived
21 Jones MP, Tack J, Van Oudenhove L, et al. Mood and anxiety disorders precede neurotrophic factor positively correlates with symptoms in functional dyspepsia. Dig
development of functional gastrointestinal disorders in patients but not in the Dis Sci 2016;61:3478–85.
population. Clin Gastroenterol Hepatol 2017;15:1014–20. 52 Gargala G, Lecleire S, François A, et al. Duodenal intraepithelial T lymphocytes in
22 Vanheel H, Carbone F, Valvekens L, et al. Pathophysiological abnormalities patients with functional dyspepsia. World J Gastroenterol 2007;13:2333–8.
in functional dyspepsia subgroups according to the Rome III criteria. Am J 53 Yuan H-­P, Li X-­P, Yang W-­R, et al. Inducible nitric oxide synthase in the duodenal
Gastroenterol 2017;112:132–40. mucosa is associated with mast cell degranulation in patients with functional
23 Talley NJ, Verlinden M, Jones M. Can symptoms discriminate among those with dyspepsia. Ann Clin Lab Sci 2015;45:522–7.
delayed or normal gastric emptying in dysmotility-­like dyspepsia? Am J Gastroenterol 54 Wallon C, Yang P-­C, Keita AV, et al. Corticotropin-­releasing hormone (CRH) regulates
2001;96:1422–8. macromolecular permeability via mast cells in normal human colonic biopsies in
24 Stanghellini V, Tack J. Gastroparesis: separate entity or just a part of dyspepsia? Gut vitro. Gut 2008;57:50–8.
2014;63:1972–8. 55 Walker MM, Aggarwal KR, Shim LS, et al. Duodenal eosinophilia and early satiety in
25 Vijayvargiya P, Jameie-­Oskooei S, Camilleri M, et al. Association between delayed functional dyspepsia: confirmation of a positive association in an Australian cohort. J
gastric emptying and upper gastrointestinal symptoms: a systematic review and Gastroenterol Hepatol 2014;29:474–9.
meta-­analysis. Gut 2019;68:804–13. 56 Wauters L, Nightingale S, Talley NJ, et al. Functional dyspepsia is associated with
26 Cherian D, Parkman HP. Nausea and vomiting in diabetic and idiopathic duodenal eosinophilia in an Australian paediatric cohort. Aliment Pharmacol Ther
gastroparesis. Neurogastroenterol Motil 2012;24:217–22. 2017;45:1358–64.
27 HG L, Weltens N, Tack J, et al. Acute anxiety and anxiety disorders are associated 57 Wauters L, Frings D, Lambaerts M, et al. Duodenal hyperpermeability and markers of
with impaired gastric accommodation in patients with functional dyspepsia. Clin inflammation are linked with gastric emptying and symptoms in functional dyspepsia
Gastroenterol Hepatol 2015;13:1584–91. patients. Neurogastroenterol Motil 2019;31.
28 Van Oudenhove L, Vandenberghe J, Geeraerts B, et al. Determinants of symptoms 58 Du L, Shen J, Kim JJ, et al. Increased duodenal eosinophil degranulation in patients
in functional dyspepsia: gastric sensorimotor function, psychosocial factors or with functional dyspepsia: a prospective study. Sci Rep 2016;6.
somatisation? Gut 2008;57:1666–73. 59 Vanheel H, Vicario M, Boesmans W, et al. Activation of eosinophils and mast cells in
29 Park S-­Y, Acosta A, Camilleri M, et al. Gastric motor dysfunction in patients with functional dyspepsia: an ultrastructural evaluation. Sci Rep 2018;8.
functional gastroduodenal symptoms. Am J Gastroenterol 2017;112:1689–99. 60 Lee K-­J, Demarchi B, Demedts I, et al. A pilot study on duodenal acid exposure and
30 Vanheel H, Farré R. Changes in gastrointestinal tract function and structure in its relationship to symptoms in functional dyspepsia with prominent nausea. Am J
functional dyspepsia. Nat Rev Gastroenterol Hepatol 2013;10:142–9. Gastroenterol 2004;99:1765–73.
31 Walker MM, Talley NJ. The role of duodenal inflammation in functional dyspepsia. J 61 Fried M, Feinle C. The role of fat and cholecystokinin in functional dyspepsia. Gut
Clin Gastroenterol 2017;51:12–18. 2002;51 Suppl 1:i54–7.
32 Vanheel H, Vicario M, Vanuytsel T, et al. Impaired duodenal mucosal integrity and 62 Keely S, Walker MM, Marks E, et al. Immune dysregulation in the functional
low-­grade inflammation in functional dyspepsia. Gut 2014;63:262–71. gastrointestinal disorders. Eur J Clin Invest 2015;45:1350–9.
33 Liebregts T, Adam B, Bredack C, et al. Small bowel homing T cells are associated with 63 Jones MP, Walker MM, Ford AC, et al. The overlap of atopy and functional
symptoms and delayed gastric emptying in functional dyspepsia. Am J Gastroenterol gastrointestinal disorders among 23 471 patients in primary care. Aliment
2011;106:1089–98. Pharmacol Ther 2014;40:382–91.
34 Cirillo C, Bessissow T, Desmet A-­S, et al. Evidence for neuronal and structural 64 Koloski N, Jones M, Walker MM, et al. Population based study: atopy and
changes in submucous ganglia of patients with functional dyspepsia. Am J autoimmune diseases are associated with functional dyspepsia and irritable
Gastroenterol 2015;110:1205–15. bowel syndrome, independent of psychological distress. Aliment Pharmacol Ther
35 Talley NJ. Editorial: moving away from focussing on gastric pathophysiology in 2019;49:546–55.
functional dyspepsia: new insights and therapeutic implications. Am J Gastroenterol 65 Walker MM, Salehian SS, Murray CE, et al. Implications of eosinophilia in the normal
2017;112:141–4. duodenal biopsy - an association with allergy and functional dyspepsia. Aliment
36 Pauwels A, Altan E, Tack J. The gastric accommodation response to meal intake Pharmacol Ther 2010;31:1229–36.
determines the occurrence of transient lower esophageal sphincter relaxations and 66 Koloski NA, Jones M, Weltman M, et al. Identification of early environmental risk
reflux events in patients with gastro-­esophageal reflux disease. Neurogastroenterol factors for irritable bowel syndrome and dyspepsia. Neurogastroenterol Motil
Motil 2014;26:581–8. 2015;27:1317–25.
37 Talley NJ. Functional dyspepsia: advances in diagnosis and therapy. Gut Liver 67 Lee KJ, Kim JH, Cho SW. Dyspeptic symptoms associated with hypersensitivity
2017;11:349–57. to gastric distension induced by duodenal acidification. J Gastroenterol Hepatol
38 van Baar ACG, Nieuwdorp M, Holleman F, et al. The duodenum harbors a broad 2006;21:515–20.
Untapped therapeutic potential. Gastroenterology 2018;154:773–7. 68 Simrén M, Vos R, Janssens J, et al. Acid infusion enhances duodenal
39 Rønnestad I, Akiba Y, Kaji I, et al. Duodenal luminal nutrient sensing. Curr Opin mechanosensitivity in healthy subjects. Am J Physiol Gastrointest Liver Physiol
Pharmacol 2014;19:67–75. 2003;285:G309–15.
40 Camilleri M. Gastrointestinal hormones and regulation of gastric emptying. Curr 69 Vanuytsel T, Karamanolis G, Van Oudenhove L, et al. Influence of ondansetron on
Opin Endocrinol Diabetes Obes 2019;26:3–10. gastric sensorimotor responses to short duodenal acid infusion in healthy volunteers.
41 Camilleri M. Leaky gut: mechanisms, measurement and clinical implications in Neurogastroenterol Motil 2011;23:226–32. e115.
humans. Gut 2019;68:1516–26. 70 Bratten J, Jones MP. Prolonged recording of duodenal acid exposure in patients with
42 Sommer F, Bäckhed F. The gut microbiota--masters of host development and functional dyspepsia and controls using a radiotelemetry pH monitoring system. J
physiology. Nat Rev Microbiol 2013;11:227–38. Clin Gastroenterol 2009;43:527–33.
43 Talley NJ, Walker MM, Aro P, et al. Non-­Ulcer dyspepsia and duodenal eosinophilia: 71 Vanheel H, Farré R, Beeckmans D, et al. Duodenal acid perfusion increases duodenal
an adult endoscopic population-­based case-­control study. Clin Gastroenterol permeability and activates the duodenogastric reflex, independently from mast cell
Hepatol 2007;5:1175–83. activation. United Eur Gastroenterol J 2017;5.
44 Friesen CA, Andre L, Garola R, et al. Activated duodenal mucosal eosinophils in 72 Potter MDE, Walker MM, Keely S, et al. What’s in a name? ’Non-­coeliac gluten
children with dyspepsia: a pilot transmission electron microscopic study. J Pediatr or wheat sensitivity’: controversies and mechanisms related to wheat and gluten
Gastroenterol Nutr 2002;35:329–33. causing gastrointestinal symptoms or disease. Gut 2018;67:2073–7.

Wauters L, et al. Gut 2019;0:1–10. doi:10.1136/gutjnl-2019-318536 9


Recent advances in clinical practice

Gut: first published as 10.1136/gutjnl-2019-318536 on 29 November 2019. Downloaded from http://gut.bmj.com/ on November 30, 2019 at Lund University Libraries. Protected by copyright.
73 Feinle-­Bisset C, Azpiroz F. Dietary and lifestyle factors in functional dyspepsia. Nat 98 Lacy BE, Saito YA, Camilleri M, et al. Effects of antidepressants on gastric function in
Rev Gastroenterol Hepatol 2013;10:150–7. patients with functional dyspepsia. Am J Gastroenterol 2018;113:216–24.
74 Staley C, Weingarden AR, Khoruts A, et al. Interaction of gut microbiota with bile 99 Tack J, Ly HG, Carbone F, et al. Efficacy of mirtazapine in patients with functional
acid metabolism and its influence on disease states. Appl Microbiol Biotechnol dyspepsia and weight loss. Clin Gastroenterol Hepatol 2016;14:385–92.
2017;101:47–64. 100 Tack J, Janssen P, Masaoka T, et al. Efficacy of buspirone, a fundus-­relaxing drug, in
75 Beeckmans D, Riethorst D, Augustijns P, et al. Altered duodenal bile salt patients with functional dyspepsia. Clin Gastroenterol Hepatol 2012;10:1239–45.
concentration and receptor expression in functional dyspepsia. United European 101 Janssen P, Harris MS, Jones M, et al. The relation between symptom improvement
Gastroenterol J 2018;6:1347–55. and gastric emptying in the treatment of diabetic and idiopathic gastroparesis. Am J
76 Beeckmans D, Riethorst D, Farré R, et al. Association between luminal bile salt Gastroenterol 2013;108:1382–91.
content and duodenal mucosal integrity in functional dyspepsia. Gastroenterology 102 Pittayanon R, Yuan Y, Bollegala NP, et al. Prokinetics for functional dyspepsia: a
2017;152:S167. systematic review and meta-­analysis of randomized control trials. Am J Gastroenterol
77 Kindt S, Tertychnyy A, de Hertogh G, et al. Intestinal immune activation in presumed 2019;114:233–43.
post-­infectious functional dyspepsia. Neurogastroenterol Motil 2009;21:832–56. 103 NICE. Available: https://​nice.​org.​uk
78 Kindt S, Van Oudenhove L, Broekaert D, et al. Immune dysfunction in patients with 104 Camilleri M, Stanghellini V. Current management strategies and emerging
functional gastrointestinal disorders. Neurogastroenterol Motil 2009;21:389–98. treatments for functional dyspepsia. Nat Rev Gastroenterol Hepatol
79 Talley NJ, Holtmann G, Walker MM, et al. Circulating Anti-­cytolethal distending 2013;10:187–94.
toxin B and Anti-­vinculin antibodies as biomarkers in community and healthcare 105 Pinto-­Sanchez MI, Yuan Y, Hassan A, et al. Proton pump inhibitors for functional
populations with functional dyspepsia and irritable bowel syndrome. Clin Transl dyspepsia. Cochrane Database Syst Rev 2017;11:CD011194.
Gastroenterol 2019;10:e00064. 106 Moayyedi P, Delaney BC, Vakil N, et al. The efficacy of proton pump inhibitors in
80 Simrén M, Barbara G, Flint HJ, et al. Intestinal microbiota in functional bowel nonulcer dyspepsia: a systematic review and economic analysis. Gastroenterology
disorders: a Rome Foundation report. Gut 2013;62:159–76. 2004;127:1329–37.
81 Sheehan D, Moran C, Shanahan F. The microbiota in inflammatory bowel disease. J 107 Potter MDE, Wood NK, Walker MM, et al. Proton pump inhibitors and suppression of
Gastroenterol 2015;50:495–507. duodenal eosinophilia in functional dyspepsia. Gut 2019;68:1339–40.
82 Qin J, Li R, Raes J, et al. A human gut microbial gene catalogue established by 108 Moayyedi P, Shelly S, Deeks JJ. Pharmacological interventions for non-­ulcer
metagenomic sequencing. Nature 2010;464:59–65. dyspepsia. In: Moayyedi P, ed. Cochrane database of systematic reviews. Chichester,
83 Shanahan ER, Zhong L, Talley NJ, et al. Characterisation of the gastrointestinal UK: John Wiley & Sons, Ltd, 2006: CD001960.
mucosa-­associated microbiota: a novel technique to prevent cross-­contamination 109 Wouters MM. Histamine antagonism and postinflammatory visceral hypersensitivity.
during endoscopic procedures. Aliment Pharmacol Ther 2016;43:1186–96. Gut 2014;63:1836–7.
84 Li G, Yang M, Zhou K, et al. Diversity of duodenal and rectal microbiota in 110 Friesen CA, Sandridge L, Andre L, et al. Mucosal eosinophilia and response to
biopsy tissues and luminal contents in healthy volunteers. J Microbiol Biotechnol H1/H2 antagonist and cromolyn therapy in pediatric dyspepsia. Clin Pediatr
2015;25:1136–45. 2006;45:143–7.
85 Zhong L, Shanahan ER, Raj A, et al. Dyspepsia and the microbiome: time to focus on 111 Potter MDE, Goodsall TM, Walker MM, et al. Dual histamine blockade for the
the small intestine. Gut 2017;66:1168–9. treatment of adult functional dyspepsia: a single centre experience. Gut 2019.
86 Kerckhoffs APM, Samsom M, van der Rest ME, et al. Lower bifidobacteria counts in doi:10.1136/gutjnl-2019-318412. [Epub ahead of print: 30 Apr 2019].
both duodenal mucosa-­associated and fecal microbiota in irritable bowel syndrome 112 Friesen CA, Kearns GL, Andre L, et al. Clinical efficacy and pharmacokinetics
patients. World J Gastroenterol 2009;15:2887–92. of montelukast in dyspeptic children with duodenal eosinophilia. J Pediatr
87 Giamarellos-­Bourboulis E, Tang J, Pyleris E, et al. Molecular assessment of differences Gastroenterol Nutr 2004;38:343–51.
in the duodenal microbiome in subjects with irritable bowel syndrome. Scand J 113 Friesen CA, Neilan NA, Schurman JV, et al. Montelukast in the treatment of duodenal
Gastroenterol 2015;50:1076–87. eosinophilia in children with dyspepsia: effect on eosinophil density and activation in
88 Shanahan ER, Shah A, Do A, et al. 146 - Duodenal Mucosa-­Associated Microbiota relation to pharmacokinetics. BMC Gastroenterol 2009;9:32.
(MAM) and Gastric Emptying: Veillonella in the Duodenal MAM Linked to slow 114 Leonard J, Marshall JK, Moayyedi P. Systematic review of the risk of enteric infection
Gastric Emptying. Gastroenterology 2018;154:S-40–40. in patients taking acid suppression. Am J Gastroenterol 2007;102:2047–56. quiz
89 Ford AC, Marwaha A, Lim A, et al. What is the prevalence of clinically significant 2057.
endoscopic findings in subjects with dyspepsia? systematic review and meta-­ 115 Tsuda A, Suda W, Morita H, et al. Influence of proton-­pump inhibitors on the luminal
analysis. Clin Gastroenterol Hepatol 2010;8:830–7. microbiota in the gastrointestinal tract. Clin Transl Gastroenterol 2015;6:e89.
90 Moayyedi P, Lacy BE, Andrews CN, et al. Acg and CAG clinical guideline: 116 Paroni Sterbini F, Palladini A, Masucci L, et al. Effects of proton pump inhibitors
management of dyspepsia. Am J Gastroenterol 2017;112:988–1013. on the gastric mucosa-­associated microbiota in dyspeptic patients. Appl Environ
91 Jackson MA, Goodrich JK, Maxan M-­E, et al. Proton pump inhibitors alter the Microbiol 2016;82:6633–44.
composition of the gut microbiota. Gut 2016;65:749–56. 117 Kim Y-­J, Chung WC, Kim BW, et al. Is Helicobacter pylori Associated Functional
92 Imhann F, Bonder MJ, Vich Vila A, et al. Proton pump inhibitors affect the gut Dyspepsia Correlated With Dysbiosis? J Neurogastroenterol Motil 2017;23:504–16.
microbiome. Gut 2016;65:740–8. 118 Mirbagheri SS, Mirbagheri SA, Nabavizadeh B, et al. Impact of microscopic
93 Ohtsu T, Takagi A, Uemura N, et al. The ameliorating effect of Lactobacillus gasseri duodenitis on symptomatic response to Helicobacter pylori eradication in functional
OLL2716 on functional dyspepsia in Helicobacter pylori-­Uninfected individuals: a dyspepsia. Dig Dis Sci 2015;60:163–7.
randomized controlled study. Digestion 2017;96:92–102. 119 Holtmann G, Talley NJ. Functional dyspepsia. Curr Opin Gastroenterol 2015;31:1.
94 Igarashi M, Nakae H, Matsuoka T, et al. Alteration in the gastric microbiota and 120 Tziatzios G, Giamarellos-­Bourboulis EJ, Papanikolaou IS, et al. Is small intestinal
its restoration by probiotics in patients with functional dyspepsia. BMJ Open bacterial overgrowth involved in the pathogenesis of functional dyspepsia? Med
Gastroenterol 2017;4:e000144. Hypotheses 2017;106:26–32.
95 Tan VPY, Liu KSH, Lam FYF, et al. Randomised clinical trial: rifaximin versus placebo 121 Pimentel M, Lembo A, Chey WD, et al. Rifaximin therapy for patients with irritable
for the treatment of functional dyspepsia. Aliment Pharmacol Ther 2017;45:767–76. bowel syndrome without constipation. N Engl J Med 2011;364:22–32.
96 Ford AC, Luthra P, Tack J, et al. Efficacy of psychotropic drugs in functional dyspepsia: 122 Meyrat P, Safroneeva E, Schoepfer AM. Rifaximin treatment for the irritable bowel
systematic review and meta-­analysis. Gut 2017;66:411–20. syndrome with a positive lactulose hydrogen breath test improves symptoms for at
97 Talley NJ, Locke GR, Saito YA, et al. Effect of amitriptyline and escitalopram on least 3 months. Aliment Pharmacol Ther 2012;36:1084–93.
functional dyspepsia: a multicenter, randomized controlled study. Gastroenterology 123 Darkoh C, Lichtenberger LM, Ajami N, et al. Bile acids improve the antimicrobial
2015;149:340–9. effect of rifaximin. Antimicrob Agents Chemother 2010;54:3618–24.

10 Wauters L, et al. Gut 2019;0:1–10. doi:10.1136/gutjnl-2019-318536

You might also like