You are on page 1of 15

Review

Gastrointestinal dysfunction in Parkinson’s disease


Alfonso Fasano, Naomi P Visanji, Louis W C Liu, Antony E Lang, Ronald F Pfeiffer

Our understanding of dysfunction of the gastrointestinal system in patients with Parkinson’s disease has increased Lancet Neurol 2015; 14: 625–39
substantially in the past decade. The entire gastrointestinal tract is affected in these patients, causing complications Morton and Gloria Shulman
that range from oral issues, including drooling and swallowing problems, to delays in gastric emptying and Movement Disorders Clinic and
the Edmond J Safra Program in
constipation. Additionally, small intestinal bacterial overgrowth and Helicobacter pylori infection affect motor
Parkinson’s Disease, Toronto
fluctuations by interfering with the absorption of antiparkinsonian drugs. The multifaceted role of the Western Hospital, University
gastrointestinal system in Parkinson’s disease necessitates a specific and detailed assessment and treatment plan. Health Network, Division of
The presence of pervasive α-synuclein deposition in the gastrointestinal tract strongly implicates this system in Neurology, University of
Toronto, Toronto, Ontario,
the pathogenesis of Parkinson’s disease. Future studies elucidating the role of the gastrointestinal tract in the
Canada (A Fasano MD,
pathological progression of Parkinson’s disease might hold potential for early disease detection and development N P Visanji PhD,
of neuroprotective approaches. Prof A E Lang MD); Division of
Gastroenterology, Department
of Medicine, Toronto Western
Introduction treatment. We conclude by discussing the next crucial Hospital, Toronto, Ontario,
Progress in the understanding of the extent and role of steps that are needed in research, treatment, and Canada (L W C Liu MD); and
gastrointestinal dysfunction in Parkinson’s disease has understanding of gastrointestinal involvement in Department of Neurology,
increased substantially in the past decade.1 Not only is Parkinson’s disease. Oregon Health and Science
University, Portland, Oregon,
the gastrointestinal system impaired from both a motor USA (Prof R F Pfeiffer MD)
and dysautonomic standpoint, but it also plays an active Synucleinopathy in the gastrointestinal tract Correspondence to:
part in the pathophysiological changes that underlie The pathological changes of Parkinson’s disease are Dr Alfonso Fasano, University of
motor fluctuations through its effects on absorption of defined by abnormal α-synuclein accumulation in the Toronto, Movement Disorders
antiparkinsonian drugs. A wealth of evidence2–4 strongly brain in characteristic Lewy bodies or Lewy neurites. Centre, Toronto Western
Hospital, Toronto, ON M5T 2S8,
implicates pathophysiological changes in the gastro- However, evidence for abnormal α-synuclein Canada
intestinal tract in the pathogenesis of Parkinson’s accumulation outside the brain, including throughout alfonso.fasano@uhn.ca
disease, and many studies5 indicate that the enteric the enteric nervous system, is growing (figure 1).
nervous system might serve as a conduit for the α-Synuclein deposition occurs in the myenteric and
propagation of α-synuclein, initiating the characteristic submucosal plexuses and mucosal nerve fibres, with a
spread of degeneration through the CNS. clear rostrocaudal gradient in deposition throughout the
Here, we review current knowledge of gastrointestinal enteric nervous system.13 A study mapping the distri-
involvement in Parkinson’s disease. First, we assess the bution of phosphorylated α-synuclein2 reported the
growing body of experimental evidence supporting the highest concentrations of enteric phosphorylated
hypothesis that the gastrointestinal tract might be the α-synuclein in the submandibular gland and lower
site of initiation of Parkinson’s disease, along with oesophagus, lower concentrations in the stomach and
evidence for the potential use of gastrointestinal small intestine, and lowest concentrations in the colon
α-synuclein deposition (from the submandibular gland and rectum. The causes of this pattern of distribution are
to the appendix and the colon) as a diagnostic biomarker. unknown, although deposition follows the pattern of
Second, we discuss the effect of gastrointestinal dys- innervation from visceromotor projection neurons.
function on clinical symptoms, including oral difficulties, These neurons originate in the dorsal motor nucleus of
swallowing problems, and constipation. As a result of the vagus nerve (DMV) and innervate the longitudinal
their frequency, these salient gastrointestinal symptoms extent of the gastrointestinal tract, with vagal innervation
are important in disease progression and constitute a predominant in the upper gastrointestinal tract and
major source of disability. We also review evidence sympathetic and pelvic outflow projections predominant
describing the deleterious effect of small intestinal in more distal regions.
bacterial overgrowth (SIBO) and Helicobacter pylori The accumulating evidence describing early enteric
infection on the absorption of antiparkinsonian drugs nervous system pathological changes in Parkinson’s
and the potential effects on the pathophysiological disease, in regions far more accessible for direct study
changes in patients with Parkinson’s disease and motor than the brain, has prompted investigations assessing
fluctuations. enteric α-synuclein as a biomarker for Parkinson’s disease.
Owing to the substantial role of the gastrointestinal Many groups, including our own, chose colonic biopsies
system in Parkinson’s disease, we believe that thorough as a practical source of tissue. We previously reviewed the
assessment and treatment of gastrointestinal dysfunction advantages and challenges of the use of colonic
in patients with the disease is essential. To this end, we α-synuclein as a diagnostic biomarker13 and concluded
have designed a practical algorithm for the assessment of that colonic α-synuclein does not provide high specificity
the gastrointestinal system in patients with Parkinson’s for the diagnosis of Parkinson’s disease.11 Our findings
disease and provide an evidence-based approach to support other reports that have shown the presence of

www.thelancet.com/neurology Vol 14 June 2015 625


Review

The appendiceal mucosa also has been proposed as a


Submandibular gland Oesophagus potential site of entry of α-synuclein because this region
• Phosphorylated α-synuclein • Phosphorylated α-synuclein in
present in postmortem tissue upper oesophagus in 0 of 8 and has the highest expression of α-synuclein in the enteric
from 28 of 28 patients with in lower oesophagus in 11 of nervous system in healthy individuals.15 Alternatively, our
Parkinson’s disease2 16 patients with Parkinson’s
• Phosphorylated α-synuclein disease8
finding9 of increased aggregated α-synuclein in the colon
present in biopsy tissue from • Lewy bodies in lower of control individuals prompted us to postulate that in
9 of 12 patients with oesophagus in 6 of 6 patients Parkinson’s disease, α-synuclein might disaggregate and
Parkinson’s disease6 with Parkinson’s disease and
3 of 8 controls9 infiltrate the CNS as soluble α-synuclein oligomers.
The distribution of synucleinopathy is associated with
gastrointestinal symptoms along the entire gastro-
Stomach intestinal tract.16 The extent of gastrointestinal dys-
• Phosphorylated α-synuclein in Colon
3 of 8 patients with Parkinson’s function, with corresponding widespread enteric nervous
• α-Synuclein in myenteric and
disease2 submucosal plexus in 10 of system synucleinopathy, could suggest that a disruption
• Lewy bodies in 2 of 7 patients
with Parkinson’s disease6
10 patients with Parkinson’s in the physiological function of α-synuclein might have a
disease and 40 of 77 controls3
• Phosphorylated α-synuclein in pivotal role in gastrointestinal dysfunction. The potential
submucosal plexus in 21 of existence of so-called strains of α-synuclein, some
29 patients with Parkinson’s
disease and 0 of 10 controls10
physiological and others with potentially deleterious
Appendix
• Appendiceal mucosa • α-Synuclein and properties, reinforces this association. However, several
phosphorylated α-synuclein reports show an absence of neuronal loss within the
had the most dense
present in mucosa in 19 of
α-synuclein expression in the
22 patients with Parkinson’s enteric nervous system in Parkinson’s disease,17 so
healthy enteric nervous
system; α-synuclein present in
disease and 11 of 11 controls11 further investigation is needed to establish whether
• α-Synuclein present in mucosa
10 of 10 individuals without
in 9 of 9 patients with
pathological α-synuclein disrupts gastrointestinal
Parkinson’s disease7
Parkinson’s disease and 0 of function in the disorder. In fact, neuronal loss in the
23 controls12 enteric nervous system might not be necessary; this
• Phosphorylated α-synuclein
present in 11 of 13 individuals finding underlines the suggested role of the DMV, a
without Parkinson’s disease with brain region that has a high level of synucleinopathy in
a possible increase with age4
Parkinson’s disease and where α-synuclein over-
expression has been shown to mediate impaired gastro-
Figure 1: Distribution of α-synuclein and phosphorylated α-synuclein in the enteric nervous system in intestinal motility in experimental parkinsonism.18
patients with Parkinson’s disease Although a clear pattern exists for the presence of
Increasing evidence suggests that abnormal α-synuclein accumulates outside the brain and throughout the enteric
synucleinopathy in regions of the gastrointestinal tract
nervous system.2–4,6–12 Adapted from Pfeiffer,1 by permission of Elsevier.
affected in Parkinson’s disease, no studies have shown a
causative link between this pathological abnormality and
α-synuclein in the colon in healthy individuals.13 Although the corresponding gastrointestinal deficit. Future work
good sensitivity for detection of α-synuclein in the colon enhancing our understanding of the association between
exists, the clear rostrocaudal gradient in α-synuclein enteric synucleinopathy and gastrointestinal dysfunction
deposition in the enteric nervous system, coupled with the might aid the development of treatments targeting
strong vagal innervation, has prompted the suggestion gastrointestinal symptoms.
that tissue from more proximal regions of the gastro-
intestinal tract might provide higher sensitivity and Oral difficulties
specificity for the diagnosis of Parkinson’s disease than Dental problems
tissue from the colon. The submandibular gland, in which Patients with Parkinson’s disease brush their teeth and
phosphorylated α-synuclein has been reported in all seek dental care less frequently than healthy individuals.19
patients with Parkinson’s disease and none of the healthy The ability of patients to brush their teeth effectively is
controls in a postmortem study, might have better hampered by decreased manual dexterity and difficulty
diagnostic potential.8 opening the jaw,20 leading to increased periodontal
The hypothesis that Parkinson’s disease might arise disease, an increased frequency of caries, and fewer
from the gut has received much attention. This idea, first remaining teeth.21 Excessive saliva (sialorrhoea) might
posited a decade ago,5 has gained attention because several change salivary pH and composition in some patients,
studies have suggested that the pathological progression of and xerostomia might impair the mouth’s self-cleaning
Parkinson’s disease might be mediated by the prion-like mechanism in others.21
properties of α-synuclein.14 The enteric nervous system is
affected early in Parkinson’s disease and the dense Drooling
innervation originating from the DMV might provide a Drooling is defined as excessive pooling of saliva in the
conduit for abnormal forms of α-synuclein to access the oral cavity due to overproduction of saliva or impaired
CNS.5 These vagal projection neurons are among the first salivary clearance, caused by swallowing difficulties or
cells in the CNS to exhibit abnormal α-synuclein deposits. inability to maintain saliva within the oral cavity. Drooling

626 www.thelancet.com/neurology Vol 14 June 2015


Review

has several negative effects: social embarrassment, poor with drooling, including disease duration, use of anti-
oral hygiene, bad breath, increased intra-oral occult depressants, hallucinations, orthostatic hypotension, and
bacteria, difficulty eating and speaking, increased risk of ageing.56 The associations with severity of motor
aspiration pneumonia, and a profound effect on quality symptoms and other motor features, and the clinical
of life.22 Furthermore, a 9·75 times increased risk of observation of increased drooling during off-times (when
respiratory infection over 1 year was documented in the effects of antiparkinsonian drugs wear off and
patients with Parkinson’s disease with daily drooling and symptoms return), suggest that impaired swallowing
silent aspiration or silent laryngeal penetration of food.23 might be the cause of drooling, rather than over-
Drooling prevalence in patients with Parkinson’s disease production of saliva.
ranges from 10% to 81% in controlled studies (table 1).29,34 Several experimental and clinical findings22 suggest
Such variability can be attributed to the absence of that salivary production is in fact reduced in many
established diagnostic criteria, the use of different types patients with Parkinson’s disease, probably as a result of
of screening questionnaires, and differences in the dopamine depletion. Accordingly, dry mouth is present
characteristics of cohorts with Parkinson’s disease.22 in more than 60% of patients with Parkinson’s disease
Several features of Parkinson’s disease are associated and might coexist with drooling in 30% of these patients.57
with drooling, including severity of motor symptoms Additional mechanisms might be the cause of drooling;
(especially dysarthria and dysphagia).54 Other motor one study29 showed that salivary production in response
features (hypomimia with unintentional mouth opening to a discrete stimulus is significantly higher in drooling
and stooped posture with dropped head) impair the
ability to maintain saliva within the oral cavity.55 Factors
Proportion of Proportion of
indirectly related to disease severity have been associated
patients with healthy controls
Parkinson’s disease

Proportion of Proportion of (Continued from previous column)


patients with healthy controls Constipation
Parkinson’s disease
Eadie and Tyrer24 61% (n=76) 13% (n=96)
Drooling Edwards et al25 29% (n=98) 10% (n=50)
Eadie and Tyrer24 50% (n=76) 5% (n=96) Singer et al41 7% (n=48) 6% (n=32)
Edwards et al25 70% (n=94) 6% (n=50) Wang et al42 71% (n=62) NA
Edwards et al26 77% (n=13) 14% (n=7) Edwards et al26 31% (n=13) 14% (n=7)
Siddiqui et al27 52% (n=44) 13% (n=24) Gonera et al43 4% (n=60) 8% (n=58)
Verbaan et al28 73% (n=420) 7% (n=150) Abbott et al44‡ 10% (n=96) 4% (n=6694)
Nicaretta et al29 10% (n=14) NA (n=8) Sakakibaraet al45 50% (n=12) 20% (n=10)
Kim et al30 26% (n=23) 4% (n=23) Chaudhuri et al46 47% (n=123) 26% (n=96)
Leibner et al31 59% (n=58) 14% (n=51) Kaye et al47 59% (n=156) 21% (n=148)
Yu et al32 46% (n=90) 13% (n=270) Verbaan et al28 22% (n=420) 2% (n=150)
Muller et al33 42% (n=207) 6% (n=175) Krogh et al48 27% (n=416) 5% (n=45)
Damian et al34 81% (n=62) 25% (n=287) Kim et al30 26% (n=23) 4% (n=23)
Picillo et al35* 16% (n=200) 2% (n=96) Savica et al49 36% (n=196) 20% (n=196)
Taste impairment Ramjit et al50 67% (n=58) 22% (n=51)
Shah et al36 27% (n=75) 1% (n=74) Yu et al32 70% (n=90) 48% (n=270)
Deeb et al37 22% (n=50) 1% (n=23) Muller et al33 39% (n=207) 14% (n=175)
Kashihara et al38 9% (n=285) 0% (n=61) Gaenslen et al51‡ 25% (n=93) 11% (n=93)
Pont-Sunyer et al39* 14% (n=109) 1% (n=107) Damian et al34 63% (n=62) 31% (n=286)
Swallowing disorders Picillo et al35* 16% (n=200) 14% (n=93)
Edwards et al25 52% (n=94) 6% (n=50) Szewczyk-Krolikowski et al52* 41% (n=490) 34% (n=176)†
Edwards et al26 77% (n=13) 14% (n=7) Pont-Sunyer et al39* 39% (n=109) 13% (n=107)
Clarke et al40 30% (n=64 4% (n=80) Schrag et al53‡§ 32% (n=7232)¶ 19% (n=40 541)¶
Siddiqui et al27 30% (n=44) 8% (n=24) 25% (n=4769)|| 15% (n=25 544)||
Verbaan et al28 55% (n=420) 19% (n=150) 20% (n=1680)** 14% (n=8305)**

Kim et al30 13% (n=23) 0% (n=23)† NA=not available. *Early disease stage, drug-naive patients.†Group comparisons
Yu et al32 28% (n=90) 21% (n=270)† were not statistically significant; all other differences between patients and
controls are significant. ‡Symptoms were assessed during the premotor phase.
Muller et al33 19% (n=207) 6% (n=175)
§Prevalence of gastrointestinal symptoms was assessed at different time intervals
Damian et al34 53% (n=60) 22% (n=290) before diagnosis: ¶2 years, ||2–5 years, and **5–10 years.
Picillo et al35* 9% (n=200) 3% (n=96)
Table 1: Prevalence of major gastrointestinal symptoms in patients with
(Table 1 continues in next column)
Parkinson’s disease reported in case-controlled studies

www.thelancet.com/neurology Vol 14 June 2015 627


Review

patients with Parkinson’s disease than in healthy secondary to basal ganglia dysfunction. However,
individuals. The association between drooling and functional neuroimaging has shown hypometabolism in
α-synuclein deposition in the submandibular glands in the supplementary motor area and the anterior cingulate
patients with Parkinson’s disease is unknown.2 cortex in these patients;64 peripheral mechanisms
including motor and sensory pharyngeal nerves might
Other aspects of oral function also play a part in oropharyngeal dysphagia.65
Impaired olfaction is well established in Parkinson’s
disease, but impairment of taste has received less Malnutrition
attention. Several studies58 have documented impaired The prevalence of malnutrition in Parkinson’s disease
taste in patients with Parkinson’s disease, with prevalence ranges from 0% to 24% and risk of malnutrition from
ranging from 9% to 27% (table 1). Age, sex, disease 3% to 60%.66 Malnutrition is an established determinant
duration, and disease stage are not associated with of health in elderly people and is linked to reduced
impaired taste, although some studies suggest that taste functional ability, quality of life, and survival in patients
is more likely to be impaired if cognitive functions are with Parkinson’s disease.67 Motor impairment (dys-
affected.58 Painful burning mouth syndrome without any phagia), fear of increased off-time, fasting associated
obvious abnormality in the oral mucosa or other with drug administration, drug-induced nausea, and
structures has been reported in patients with Parkinson’s anorexia can all affect food intake. Female sex, ageing,
disease and, in some instances, might be induced by loss of appetite, depression, apathy, and loss of olfaction
levodopa treatment.59 or taste might also contribute to malnutrition.
Dyskinesias contribute to weight loss by increasing
Swallowing disorders energy expenditure,68 and low bodyweight is associated
Oropharyngeal and oesophageal dysphagia are frequently with an increased risk of dyskinesias, creating a vicious
reported in patients with Parkinson’s disease, with a circle.68,69 Administration of levodopa (normalised for
prevalence ranging from 9% to 77% (table 1). In a meta- bodyweight and adjusted for age, sex, and disease
analysis,60 the pooled prevalence was estimated to be 35% severity) has been associated with impaired nutritional
for subjective oropharyngeal dysphagia; however, status in a dose-dependent manner.70 A direct effect of
because many patients are unaware of symptoms, levodopa on fat metabolism, skeletal muscle glucose
objective testing resulted in a higher prevalence of 82%.60 uptake, and hormones (such as prolactin) has also been
Pharyngeal dysfunction and oropharyngeal transit postulated.71 A cumulative levodopa dose is purportedly
abnormalities increase the risk of aspiration, thus associated with nutritional deficiency through changes
contributing to risk of upper respiratory tract infection in homocysteine, vitamin B6, and B12 levels.72
and pneumonia.61 Aspiration is present in 15–56% of Conversely, weight gain is associated with treatment
patients with Parkinson’s disease and silent aspiration in with dopamine agonists and neurosurgical procedures,
15–33% of patients.62 such as deep-brain stimulation of the subthalamic
Dysphagia typically emerges in more advanced nucleus, possibly due to effects on limbic areas.73
Parkinson’s disease, but it can develop early, occasionally
even as the presenting feature.63 Oropharyngeal Stomach problems
dysphagia is often attributed to bradykinesia and rigidity Motility disorders
Recognition of impaired gastric emptying in Parkinson’s
disease is growing; prevalence ranges from 70% to 100%,
although not all affected individuals have symptoms.74
Gastroparesis can be present in both early and advanced
Parkinson’s disease, with delays in gastric emptying
more likely to be associated with solid meals.75 Nausea,
vomiting, early satiety, excessive fullness, bloating, and
abdominal distension characterise gastroparesis.76
Because levodopa is absorbed in the small intestine,
gastroparesis might result in delayed or complete loss of
benefit of the levodopa dose (figure 2).
The pathophysiological abnormalities of gastroparesis
in Parkinson’s disease probably include central and
enteric components. The DMV is involved early in the
course of disease5 and, in 6-hydroxydopamine-lesioned
animals, delayed gastric emptying is prevented by
Figure 2: Delay in gastric emptying
vagotomy.77 However, involvement of the enteric nervous
Photograph taken during gastroscopy. Arrow points to a carbidopa tablet system might occur even earlier in Parkinson’s disease,78
remaining intact in a patient’s stomach about 1·5 h after intake. suggesting that peripheral factors also contribute to

628 www.thelancet.com/neurology Vol 14 June 2015


Review

gastroparesis in these patients. The neuropeptide ghrelin


has a role in the modulation of gastric motility; ghrelin is Weight loss, iron or
vitamin deficiency
secreted when the stomach is empty, thus increasing
gastrointestinal motility in preparation for ingestion of
food. Abnormal ghrelin function might therefore be
Malabsorption
implicated in gastroparesis in Parkinson’s disease,
suggesting that ghrelin or its receptor might be potential
treatment targets.79
Parkinson’s disease

Helicobacter pylori infection


The spiral Gram-negative bacterium H pylori is a highly
prevalent risk factor for peptic ulcers and gastric cancer. A Parkinson’s disease
H A G drugs A H
chronic systemic inflammatory response to H pylori could
be a common determinant in the onset, evolution, and
outcome of many neurological diseases, including Helicobacter pylori BC
Slow gastrointestinal Small intestinal
Parkinson’s disease. The authors of a Danish population- B
motility bacterial overgrowth
based study80 reported that a diagnosis of H pylori gastritis
was associated with about a 45% increased risk of
Parkinson’s disease. The peripheral immune response, Proton-pump inhibitors D

triggered by H pylori, might disrupt the blood–brain


barrier, thus promoting microglia-mediated inflammation
E
and autoimmune neurotoxic effects.81 However, the Motor fluctuations F
relevance of this association is unclear because many
shared risk factors for the development of H pylori Figure 3: The complex association between Helicobacter pylori, small intestinal bacterial overgrowth, and
infection and Parkinson’s disease exist, and the prevalence Parkinson’s disease
of H pylori in Parkinson’s disease is similar to that in the (A) H pylori and small intestinal bacterial overgrowth (SIBO) might affect the progression of Parkinson’s disease
through induction of the peripheral immune response, disruption of the blood–brain barrier, and mediation of
general population.82
neuroinflammation.81,84 The dotted line represents no clear evidence. (B) H pylori and SIBO negatively affect
Strong evidence exists for a biological contribution of gastrointestinal motility by means of various mechanisms—eg, reduction in the production of ghrelin.85 (C) Slow
H pylori to motor fluctuations in Parkinson’s disease, motility might lead to increased prevalence of SIBO.86 (D) The use of proton-pump inhibitors and hypochlorhydria
owing to interference in the absorption of levodopa related to atrophic gastritis increases the likelihood of concomitant SIBO in patients infected with H pylori.
(E) H pylori can affect motor fluctuations by means of gastric hypochlorhydria (levodopa is highly soluble in an acidic
(figure 3).83 Accordingly, in a diary-based study,87 H pylori environment); the presence of adhesins on the bacteria surface that directly bind to levodopa; local production of
infection was associated with an increased delay in onset reactive oxygen species, which inactivate levodopa; levodopa acting as a nutrient for bacterial growth; impairment
of action of levodopa and decreased on-time (time during of stomach motility, leading to a prolonged transit time and exposure of the drug to bacteria and enzyme activity;
which symptoms are adequately controlled). The findings and disruption of the mucosa in the small intestine, which is the main site of levodopa absorption.83 (F) SIBO can
affect motor fluctuations through levodopa malabsorption owing to changed chyme composition as a result of
of a small clinical study88 did not lend support to these mucosal injury (loss of activity of brush-border disaccharidases) or the bacterial fermentation of sugars and
results, but patients infected with H pylori were treated deconjugation of bile acids (thus leading to the production of lithocholic acid, which is directly toxic to
with higher doses of levodopa than those not infected. enterocytes).82 (G) Parkinson’s disease affects motor fluctuations through its effects on gastric emptying
Studies investigating levodopa pharmacokinetics did not (like H pylori and SIBO). (H) H pylori and SIBO are common causes for malabsorption of important nutrients.
find differences between patients with or without H pylori
infection.83,89 This inconsistency in clinical and pharma- inhibits retrograde migration of bacteria from the colon
cokinetic findings could result from the tightly controlled into the ileum.90 A prospective study86 provided evidence
conditions of the levodopa challenge in pharmacokinetic for an increased prevalence of SIBO in patients with
studies, which might not reflect the patient’s own Parkinson’s disease that was associated with the Hoehn
alimentary habits. Nevertheless, the strongest evidence and Yahr stage and motor score from the unified
supporting a direct role of H pylori in the pathophysiological Parkinson’s disease rating scale. Another study focusing
changes associated with motor fluctuations comes from on patients with fluctuating Parkinson’s disease82
H pylori eradication studies. showed a similar increased prevalence (55% vs 20% in
healthy controls), whereas an uncontrolled study in 103
Small intestinal bacterial overgrowth consecutive patients noted a lower prevalence (25%),
In healthy people, intrinsic mechanisms control the probably due to patients having less severe disease and
number and composition of small intestine microbiota: exclusion of patients taking proton-pump inhibitors,
gastric acid destroys many bacteria entering the which increase the risk of SIBO in patients with
stomach; biliary and pancreatic secretions limit bacterial Parkinson’s disease.84
growth; the migrating motor complex (which has the Patients with Parkinson’s disease with SIBO have been
housekeeping function of gastrointestinal motility) reported to have more severe motor fluctuations (off-
reduces luminal growth potential; the intestinal mucus time, delayed on-time, and no on-time) than those
traps and fights bacteria; and the ileocaecal valve without SIBO.82 This study also controlled for concurrent

www.thelancet.com/neurology Vol 14 June 2015 629


Review

H pylori infection, which was linked to wearing-off population-based study reported that risk of
episodes and contributed with SIBO to severity of off- development of Parkinson’s disease increases with
time, absence of on-episodes (dose failure), and presence constipation severity (hazard ratios ranged from 3·3 to
of unpredictable fluctuations.82 Although the results of a 4·2).96 Not only is the risk increased in individuals with
more recent study that adjusted for disease duration84 did constipation, but constipation is also emerging as one
not confirm an association with motor fluctuations, a of the earliest features of autonomic dysfunction in
significant association between SIBO and worse motor Parkinson’s disease, developing as early as 15·3 years
function was identified. before motor features.97 Thus, detection of constipation
Any causative link between SIBO and Parkinson’s might have potential sensitivity as a clinical biomarker
disease severity is difficult to ascertain. The peripheral of prodromal Parkinson’s disease.
inflammatory state induced by SIBO might contribute to The mechanisms underlying constipation in
increased intestinal permeability, thus promoting the Parkinson’s disease are unknown. Reduced neuronal
translocation of bacteria and endotoxins across the density in the myenteric ganglion can be present in
intestinal epithelium, triggering microglial activation patients with severe chronic constipation.98 However,
and exacerbating the neurodegenerative process.84 There- an absence of neuronal loss in the myenteric plexus in
fore, SIBO might create a proinflammatory environ- Parkinson’s disease has been reported;99 in another
ment,91 as reported in newly diagnosed patients with study,100 there was evidence of phosphorylated
Parkinson’s disease, and supported by a study that α-synuclein in four of five colonic biopsy samples from
quantified local inflammation and enteric glial reaction individuals with Parkinson’s disease and concurrent
in gastrointestinal biopsies of patients with Parkinson’s chronic constipation, with no staining evident in those
disease.92 Nonetheless, a clear negative effect of SIBO on with Parkinson’s disease or chronic constipation alone,
levodopa absorption is present (figure 3). SIBO might but these samples did not include investigation of the
change chyme composition as a result of mucosal injury myenteric plexus.
or the bacterial fermentation of sugars and deconjugation Constipation is a common adverse effect of many
of bile acids.82 SIBO might also impair drug absorption Parkinson’s disease drugs, particularly anticholinergics
as a result of inflammation or partial metabolism of and dopamine agonists. Nevertheless, delayed colonic
levodopa. SIBO might contribute to reduced gastric transit has been reported in patients with Parkinson’s
emptying through inflammatory effects on entero- disease independent of drugs.101 Patients might also have
chromaffin-like cells.93 As for H pylori, evidence sup- dyssynergic defecation, which is characterised by the
porting a direct role of SIBO in the pathophysiological inability, or paradoxical increase, of the pelvic floor
changes associated with motor fluctuations comes from muscles to relax during attempted defecation. Although
eradication studies (see below). dysfunction of the sacral parasympathetic nucleus and
Because the intestinal microbiota influence the immune pelvic ganglia is implicated in Parkinson’s disease, the
system and the absorption of nutrients, drugs, and toxic underlying pathogenesis of dyssynergic defecation
compounds, a study was done to investigate the faecal might involve dopaminergic dysfunction, because acute
microbiome of 72 patients with Parkinson’s disease and administration of apomorphine improves symptoms of
matched healthy controls.94 An under-representation of dyssynergic defecation in a subset of patients with
the Prevotella-associated gut microbiome enterotype and Parkinson’s disease.26 Dyssynergic defecation has also
an abundance of Lactobacillaceae, Verrucomicrobiaceae, been reported in 25–30% of patients with idiopathic
Bradyrhizobiaceae, and Clostridiales incertae sedis IV chronic constipation and can be improved with bio-
were independently associated with Parkinson’s disease. feedback (pelvic floor rehabilitation training), suggesting
Additionally, Enterobacteriaceae abundance was that maladaptive behaviour rather than an irreversible
associated with more severe axial motor symptoms. The structural defect might explain some cases.102
scarcity of Prevotellaceae is related to increased gut per-
meability and lower concentrations of vitamins such as A practical algorithm for management
thiamine and folate, conditions previously reported in All patients with Parkinson’s disease with gastrointestinal
Parkinson’s disease.94,95 A decreased abundance of Prevo- symptoms or with a suboptimum response to
tellaceae and an increased abundance of Lactobacillaceae dopaminergic drugs (possibly related to poor absorption)
have also been associated with reduced concentrations of should be assessed and treated (figure 4). Identification
the gut hormone ghrelin.94 of modifiable factors is a crucial first step in the manage-
ment of these patients. In some cases, Parkinson’s
Constipation disease treatment should be optimised—eg, because
Constipation is the most common gastrointestinal drooling is common during off-times, treatment of
symptom in Parkinson’s disease, reported in 80–90% of fluctuations should first be addressed, although this
patients (table 1), and in view of its emergence long must be weighed against the fact that many anti-
before the onset of motor symptoms, is an especially parkinsonian drugs can cause or exacerbate gastro-
noteworthy gastrointestinal feature of the disease. A intestinal symptoms. A multidisciplinary approach is key

630 www.thelancet.com/neurology Vol 14 June 2015


Review

to the success of gastrointestinal management because has been reported in about 67% of patients with
most of these conditions will need the coordinated Parkinson’s disease on the basis of oesophageal
intervention of several health professionals, such as manometry studies.106 In patients who are unable to
speech pathologists, dietitians, neurologists, and gastro- tolerate an oesophageal manometry study, barium
enterologists. This approach further emphasises the swallow can be used. Endoscopy should be considered if
importance of non-pharmacological and rehabilitative structural or inflammatory lesions are suspected.
management. In an evidence-based review,107 preliminary evidence
was identified for the effectiveness of expiratory muscle
Dental problems and drooling strength training and video-assisted swallowing therapy
Because individuals with Parkinson’s disease who have for dysphagia. The role of dopaminergic drugs and
removable dentures might have difficulties mani- deep-brain stimulation surgery is controversial.135–137 No
pulating them, dental implants might be preferable and studies have assessed the use of enterokinetic drugs in
should be considered earlier, rather than later, in the oesophageal dysphagia; thus, treatment should be
course of disease.110 Treatment of drooling includes dictated by the underlying dysmotility.
behavioural interventions, including changes of head
position, use of swallow timers or chewing gum,111 and Malnutrition
pharmacological approaches (table 2). For patients with Parkinson’s disease who have
substantial or persisting weight loss, assessment for an
Swallowing problems alternative medical cause should be undertaken. Mal-
Patients with Parkinson’s disease and dysphagia should nutrition and its metabolic effects (vitamin deficiencies
be considered for assessment of both oropharyngeal and and sarcopenia) should not be overlooked. Nutritional
oesophageal dysphagia, guided by clinical history and intervention (individualised dietetic advice) might
presentations. Videofluoroscopy is the gold standard in contribute to improved quality of life.138
the assessment of oropharyngeal dysfunction in patients
with Parkinson’s disease105 and an oesophageal mano- Gastroparesis
metry study is the most accurate and sensitive test to The best clinically validated assessment for gastro-
identify oesophageal dysmotility. Oesophageal dysphagia paresis is measurement of the gastric emptying time

Patients with Parkinson’s disease with gastrointestinal symptoms or suboptimal response to dopaminergic drugs

Occult infections Motility problems

Helicobacter pylori Small intestinal bacterial Dysphagia Gastroparesis Constipation


overgrowth

Assessments Assessments Assessments Assessments Assessments


• Urea breath test103 • Lactulose breath test82 • Oropharyngeal • Technetium-99-labelled • Outlet
• Stool antigen test103 • Glucose breath test82 • Videofluorography105 gastric emptying study108 • Anorectal manometry
• Serology • Jejunal aspirate90 • Oesophageal • Balloon expulsion test
• Gastroscopy83,89 • Oesophageal Treatments • Defecography109
Treatments manometry study106 • Dopamine antagonists* • Transit
Treatments • Antibiotic cycles90 • Upper gastrointestinal • 5-HT4 agonists • Radio-opaque marker
• Eradication therapies104 series barium • Macrolide antibiotics study

Treatments Treatments
• Oropharyngeal • Outlet
• Expiratory muscle • Biofeedback102
strength training • Botulium neurotoxin
• Video-assisted injection to anal
swallowing therapy107 sphincter101
• Transit102
• Fibre
• Laxatives (osmotic,
stimulant)
• Prokinetic drugs
• Secretagogues

Figure 4: A practical algorithm for the assessment of patients with Parkinson’s disease with gastrointestinal dysfunction
*All dopamine antagonists used for their anti-enteric effects (eg, metoclopramine) are contraindicated in Parkinson’s disease with the exception of domperidone.

www.thelancet.com/neurology Vol 14 June 2015 631


Review

during the course of 4 h with a standard meal labelled needs diligent monitoring.139 The 5-HT4 receptor
with Technetium-99.108 Treatment of gastroparesis in agonists cisapride and tegaserod are effective, but have
Parkinson’s disease is challenging because levodopa been withdrawn from the market because of cardiotoxic
might delay gastric emptying. A widely used dopamine effects and are available only with restricted access in
antagonist, metoclopramide, is contraindicated in some countries. Prucalopride is a newer 5-HT4 agonist
Parkinson’s disease because it crosses the blood–brain with no documented cardiotoxic effects and has been
barrier and exacerbates motor dysfunction. studied in patients with chronic constipation; however,
Domperidone, another dopamine antagonist, does not its usefulness in gastroparesis has not been investigated.
cross the blood–brain barrier, but is not universally The macrolide antibiotic erythromycin, although
available and increased risk of cardiac arrhythmia effective, is not suitable for chronic use owing to drug

Dose Evidence in patients with Outcomes Side-effects


Parkinson’s disease
Systemic anticholinergics
Glycopyrrolate 1–2 mg twice or three times 4-week randomised double-blind, Significant change in No difference from placebo
daily placebo-controlled crossover trial in sialorrhoea scoring scale (blood–brain barrier not
23 patients112 crossed); behavioural changes
reported in children and young
adults with cerebral palsy;113
could cause peripheral
side-effects (eg, constipation)
Topical anticholinergics
Sublingual atropine 0·5 mg twice daily (eg, 1 drop 1-week open-label study in six Improvement in objective Can cross the blood–brain
of 1% atropine solution) patients114 and subjective measures barrier, producing the same
side-effects as systemic
administration, including
confusion114
Sublingual ipratropium 21–42 μg (1–2 sublingual 5-week randomised double-blind, No improvement in objective No difference from placebo
bromide sprays) up to four times daily placebo-controlled, crossover study in evaluation (primary (blood–brain barrier not
17 patients115 endpoint) but improvement crossed)
in subjective ratings
Tropicamide Intra-oral films up to Single-dose pilot study with a A non-significant No difference from placebo
3 mg, one dose randomised double-blind, improvement was seen in
placebo-controlled, crossover design in the 0·3 mg and 1 mg dosage
12 patients116 groups
Systemic alpha-2 agonists
Clonidine 0·15 mg per day 12-week double-blind, Significant improvement of Diurnal somnolence, dizziness,
placebo-controlled study in the frequency of clearing and dry mouth
32 patients117 saliva
Systemic alpha-1 agonists
Modafinil 100 mg per day A study in patients with Parkinson’s Reduction in drooling Positive effect on drooling
disease and Hoehn and Yahr grade 2–3 severity score and might be related to the
with moderate to severe drooling;118 patient-reported improvement of dysphagia22
study type not available improvement
Botulinum neurotoxin serotype A and B
Onabotulinumtoxin A Parotid gland 5–50 U; A case series;119 three open-label Similar positive outcomes for Safe; only minimum side-
submandibular gland 5 U studies;120–122 an open-label case-control botulinum neurotoxin A effects reported—dryness of
study;123 a randomised (abobotulinumtoxin A) and mouth and increased saliva
placebo-controlled study;124 and botulinum neurotoxin B, but viscosity, which might depend
a randomised double-blind, a faster effect after on the distribution of
placebo-controlled study125 botulinum neurotoxin B, botulinum neurotoxin in the
Abobotulinumtoxin A Parotid gland 75–146·2 U; A case series;127 two randomised probably in view of an major salivary glands
submandibular gland 78·7 U double-blind, placebo-controlled increased affinity of
studies;128,129 and a randomised rimabotulinumtoxin B for
double-blind, crossover trial compared autonomic terminals126
with rimabotulinumtoxin B126
Rimabotulinumtoxin B Parotid gland 500–2000 U; Two open-label studies;130,131 three
submandibular gland 250 U randomised double-blind,
placebo-controlled studies;132–134 and
a randomised double-blind, crossover
trial compared with
abobotulinumtoxin A126

Table 2: Pharmacological treatments for drooling in patients with Parkinson’s disease

632 www.thelancet.com/neurology Vol 14 June 2015


Review

interactions and QT prolongation, and studies have probability of H pylori infection in the patient population;
suggested that azithromycin might be a preferable the test can also remain positive long after H pylori
alternative.140 Further research is needed to assess the eradication. In patients who present with foregut
clinical efficacy of nizatidine, a selective histamine symptoms, gastroscopy is helpful to detect an active
H2 receptor agonist,141 and ghrelin agonists.142 H pylori infection and to quantify the severity of gastritis,
Botulinum neurotoxin injections into the pyloric which is proposed to affect levodopa absorption.83,89 The
sphincter have been anecdotally reported to ameliorate urea breath test and stool antigen test can be used to
gastroparesis in patients with Parkinson’s disease, but assess continued infection after eradication therapy.103
extensive investigation in patients without Parkinson’s The positive effect of eradication of a gastrointestinal
disease but with gastroparesis has not shown a clear infection on the promotion of levodopa absorption was
benefit.143 Improvement in gastric emptying after deep- originally described in one patient with Parkinson’s
brain stimulation of the subthalamic nucleus has been disease with duodenitis caused by Strongyloides
reported,144 but needs further study. Finally, although not stercoralis.146 The effect of H pylori eradication on motor
studied in patients with Parkinson’s disease, gastric fluctuations has been confirmed in a double-blind
electrical stimulation might be effective in refractory study;89 the improvement in motor fluctuations was
cases.145 greater after 3 months than in shorter-term follow-up,
suggesting a time-dependent decrease in H pylori-related
Infections inflammatory changes in gastrointestinal mucosa.87
Helicobacter pylori Several meta-analyses and systematic reviews104 have
Non-invasive tests for H pylori infection include the urea yielded inconsistent conclusions as to which first, second,
breath test, the stool antigen test, and serological tests. or third-line combination regimen is the most effective to
The positive predictive value of H pylori serological tests eradicate infection. The triple therapy regimen (proton-
is highly variable and dependent on the pre-test pump inhibitors, amoxicillin 1 g [metronidazole 500 mg if

Rationale with regard to gastrointestinal Highest level of evidence Advantages Disadvantages


dysfunction
Orally dissolving levodopa Easily administered in patients with Single-dose, double-blind, Can be taken without water Not a true sublingual preparation;
swallowing difficulties (orally double-dummy, crossover study absorbed in the gastrointestinal tract
disintegrating levodopa is swallowed with in 20 patients 156
rather than through the oral mucosa; only
saliva) a small pilot study is available with no
significant group differences156
Levodopa methyl ester The liquid formulation limits the effect of Randomised double-blind, Improvement of afternoon dose No improvement of total off-time
(melevodopa) delayed gastric emptying double-dummy, controlled failures (increased bioavailability and besides a statistical trend (p=0·07)157
effervescent tablets parallel-group trial in decreased time to onset) compared
221 patients157 with standard levodopa163,164
Dispersible levodopa The liquid formulation limits the effect of Open-label crossover trial in Fast onset of action Few good-quality studies, pulsatile and
delayed gastric emptying eight patients158 erratic delivery, and poor solubility in
water
Levodopa-carbidopa Levodopa is delivered into the jejunum; Randomised double-blind, Significant reduction of off-episodes Levodopa undergoes competition with
intestinal gel bypassing the stomach ensures a rapid and double-dummy trial and disabling dyskinesias; simplification dietary aminoacids for transport across
reliable onset of action in 66 patients162 of oral treatment, with reduction of the blood–brain barrier;166 high rate of
dopaminergic side-effects165 percutaneous endoscopic gastrostomy
complications; not approved for 24 h
continuous infusion
Apomorphine Subcutaneous delivery of a strong Double-blind, single-dose, Effectively aborts off-episodes as a Few good-quality studies for continuous
dopamine agonist, bypassing the crossover trial in 62 patients160 rescue treatement;167 continuous infusion; side-effects might be both local
gastrointestinal system; can be given either infusion reduces motor fluctuations (subcutaneous nodules) and systemic
with intermittent (pen-jet) or continuous and dyskinesias168 (nausea, hypotension, psychosis)
infusion (micro-pump)
Rotigotine patch Subcutaneous delivery of a dopamine Randomised double-blind, Improvement of non-motor High rate of reactions at the application
agonist, bypassing the gastrointestinal controlled study of the symptoms; reduced incidence of site
system rotigotine patch vs placebo and psychiatric complications;169 good
ropinirole in 561 patients159 strategy when oral therapy cannot be
used (eg, pre-surgery and post-surgery)
Deep-brain stimulation of Direct targeting of basal ganglia Randomised controlled study of Significant reduction of off-episodes Surgical risk (bleeding); postoperative
the subthalamic nucleus dysfunction deep-brain stimulation vs best and disabling dyskinesias; reduction of depression and suicide;
medical therapy in oral medications mainly possible with stimulation-induced side-effects
251 patients161 deep-brain stimulation of the (hyphophonia)
subthalamic nucleus but not the globus
pallidus interna161,170

Table 3: Treatments designed to account for the presence of gastrointestinal dysfunction in patients with Parkinson’s disease

www.thelancet.com/neurology Vol 14 June 2015 633


Review

allergic to penicillin], and clarithromycin 500 mg twice Eradication of SIBO improved motor fluctuations (off-
daily for 7 days) has been proposed as the first-line time and delayed on-time) without affecting the
treatment for H pylori eradication103 and has also been pharmacokinetics of levodopa in an open-label study of
used in patients with Parkinson’s disease. A meta- rifaximin 200 mg three times a day for 1 week.82 No
analysis147 showed that lengthening the treatment duration adverse effects related to the antibiotic therapy were
(7 days vs 10 days vs 14 days) increases the eradication rate reported and 14 of 18 patients did not have SIBO after
with only a marginal increase in adverse events between 1 month. However, in view of the persistence of
7 days and 14 days, but the eradication rate did not differ underlying predisposing factors (ie, the presence of
between 10 days and 14 days. The treatment course of Parkinson’s disease), the rate of SIBO relapse at
choice will depend on regional pharmacotherapy 6 months was, unsurprisingly, high, occurring in 6 of
availability, local H pylori antibiotic resistance profile, and 14 individuals compared with 22 of 80 in another study
patient tolerability. Of note, quinolone-based therapy in patients without Parkinson’s disease.149
should be used with caution owing to the risk of adverse Many different antibiotic regimens have been advocated
CNS effects in elderly people.148 for SIBO, including ciprofloxacin, metronidazole, neo-
mycin, norfloxacin, and doxycycline, with no consensus
Small intestinal bacterial overgrowth about the most efficacious dose or duration of treatment.90
Although the gold standard for the diagnosis of SIBO is The common antibiotic treatment cycle is 1–2 weeks every
colony counts of cultured jejunal aspirate, this method is 1–3 months based on clinical response. When available,
invasive and its diagnostic reliability is highly variable.90 rifaximin (a non-absorbed rifamycin analogue) has the
The lactulose breath test and the glucose breath test, most favourable tolerability.
measuring breath hydrogen after oral administration of
lactulose or glucose, are widely used in clinical studies of Constipation
SIBO. Some patients with SIBO harbour microbiota that In patients with chronic constipation, a thorough history
produce methane gas rather than hydrogen (eg, and digital rectal examination is helpful to differentiate
Staphylococcus aureus, Streptococcus viridans, Enterococci, colonic transit abnormality versus dyssynergic de-
Serratia, or Pseudomonas). Thus, use of the glucose fecation.102,109 The usefulness of the radio-opaque marker
breath test and the lactulose breath test to measure both study for colonic transit measurement, and of anorectal
hydrogen and methane has been suggested to increase manometry, the balloon expulsion test, and defecography
the accuracy of diagnosis of SIBO.82 have been described in patients with idiopathic chronic
constipation.109 However, none of these motility tests
have been validated in patients with Parkinson’s disease.
Although constipation is the most common gastro-
A intestinal symptom in patients with Parkinson’s disease,
trials to assess efficacy of various drugs have been scarce.
D Results from an open-label observational study in ten
patients with Parkinson’s disease showed that an osmotic
B laxative (macrogol 3350) was efficacious.150 A 2-week
randomised, placebo-controlled trial in 54 patients showed
that lubiprostone, a secretagogue and chloride channel
E activator, is efficacious without substantial adverse effects,
although nausea is common.151,152 Cisapride and tegaserod
C showed promise in the treatment of constipation in
Parkinson’s disease, but have been withdrawn because of
concerns about cardiotoxic effects.153 Prucalopride, an
enterokinetic drug with the same mechanism of action as
cisapride and tegaserod, and linaclotide, a guanylate
cyclase C agonist luminally active and minimally absorbed
gastrointestinal secretagogue, have not been studied in
Figure 5: Treatment options developed to bypass gastrointestinal patients with Parkinson’s disease.154 In view of the few
dysfunction in patients with Parkinson’s disease
trials in patients with Parkinson’s disease with
(A) Liquid levodopa formulations, in the form of dispersible levodopa or soluble
melevodopa, might enable more rapid onset of drug action than standard constipation and transit dysfunction, we recommend use
formulations.156–158 Parenteral administration routes including (B) the rotigotine of the same treatment algorithm as in patients with
patch and (C) subcutaneous apomorphine are marketed in many countries.159,160 idiopathic chronic constipation.102,109 In patients with
(D) Deep-brain stimulation of the subthalamic nucleus allows a reduction in
dyssynergic defecation,155 although botulinum neurotoxin
levodopa equivalent daily dose of about 50%, which might decrease
gastrointestinal related side-effects.161 (E) The stomach can be bypassed by injections into the anal sphincter have been recom-
intrajejunal infusion of a levodopa-carbidopa gel through a tube inserted into a mended,101 further study is needed before recommending
percutaneous endoscopic gastrostomy.162 this approach as standard practice.

634 www.thelancet.com/neurology Vol 14 June 2015


Review

Bypassing the gastrointestinal tract


Gastrointestinal dysfunction clearly plays a part in the Search strategy and selection criteria
development of motor fluctuations and dyskinesia in We identified references for this Review from searches of our
Parkinson’s disease. Therefore, treatment options partly personal files and textbooks. We also searched PubMed for
or completely bypassing the gastrointestinal tract have papers published in English from Jan 1, 1965, to Jan 26, 2015,
been developed (table 3 and figure 5). Because with the search terms “gastrointestinal dysfunction”, “weight
absorption of levodopa in tablet form is affected by loss”, “dysphagia”, “gastroparesis”, “gastric emptying”, “small
unpredictable gastric emptying time (figure 2), many intestine”, “colon”, “anorectal”, “dental”, “drooling”, “taste”,
liquid levodopa formulations have been developed and “burning mouth”, “dry mouth”, “nutrition”, “helicobactor
are commercially available (table 3). Another alternative pylori”, “small intestinal bacterial overgrowth”,
is orally dissolving levodopa (swallowed with saliva), but “constipation”, and “enteric synuclein” coupled with the term
levodopa absorption still depends on the function of the “Parkinson’s disease”. We selected the final list of references
proximal intestine, and thus requires an intact small on the basis of specific relevance to this Review and focused
bowel.171 Therefore, parenteral routes have been on advances from the past 10 years.
investigated (table 3 and figure 5). The stomach can be
bypassed with functional neurosurgery, rotigotine trans-
dermal patch, apomorphine subcutaneous infusion or approaches, although the association between infection
injection, or a percutaneous endoscopic gastrostomy status and motor function or response to treatment is
with jejunal tube for intrajejunal levodopa infusion,162 not straightforward and, in view of their high incidence
making treatment possible in patients with severe in the ageing population, H pylori and SIBO might be
swallowing problems. Many patients nowadays are unrelated to Parkinson’s disease. Future studies with
started on parenteral treatment using rotigotine. improved diagnostic capabilities should allow an
However, monotherapy with dopamine agonists often accurate estimation of prevalence. An improved
does not suffice when motor signs progress. Thus, novel understanding of the interaction between H pylori and
levodopa formulations are already under development: SIBO is needed because the likelihood of concomitant
microspheres for drug delivery and esters or alkyl esters SIBO is very high in patients infected with H pylori and
for transdermal, intranasal, subcutaneous, or intra- some effects related to H pylori eradication might be
muscular administration, and microenemas for rectal caused by concomitant SIBO eradication.82,89
administration. Additionally, because only 10% of patients with
continuing H pylori infection have signs of atrophic
Conclusions gastritis, future studies stratifying participants in
Understanding of the pivotal role of the gastrointestinal different subgroups should be undertaken.173 The roles
system in the pathophysiology and possibly also the of hybrid and super-optimised H pylori eradication
cause of Parkinson’s disease has grown substantially therapies and probiotics also need to be explored.
during the past decade. Although some progress has Research into alternative routes of administration of
been made in the development of effective treatments levodopa formulations that bypass the gastrointestinal
for gastrointestinal dysfunction in these patients, much tract are underway, and potential innovations include
remains to be accomplished. More effective treatment microspheres for drug delivery and esters or alkyl esters
modalities for dysphagia are sorely needed. Problems for transdermal, intranasal, subcutaneous, and intra-
with potential cardiotoxic effects have impeded the muscular administration, and microenemas for rectal
introduction of effective medical treatments for application. In the coming years, our understanding of
gastroparesis. However, newer drugs, such as the ghrelin gastrointestinal dysfunction in Parkinson’s disease will
agonist relamorelin, hold great promise if positive hopefully continue to accelerate and lead to improved
results in patients with diabetic gastroparesis can be treatment strategies for patients.
replicated in patients with Parkinson’s disease.172 Contributors
Relamorelin is being assessed in a multicentre clinical All authors contributed to the writing of the first draft of the manuscript
trial in patients with Parkinson’s disease and chronic and editing thereafter.
constipation, and effects on gastroparesis will also be Declaration of interests
assessed in this study (ClinicalTrials.gov, NCT01955616). AF has received honoraria from UCB, Medtronic, Boston Scientific,
Abbvie, and Teva Canada. LWCL has received honoraria from Abbvie,
Although various drugs are available for the treatment of Actavis Canada, Forest Laboratories Canada, and Takeda Canada. AEL
slow-transit constipation, more effective prokinetic has served as an adviser for Abbvie, Allon Therapeutics, Avanir
agents, such as relamorelin, are still needed. Additionally, Pharmaceuticals, Biogen Idec, Boerhinger Ingelheim, Ceregene, Lilly,
more research is needed into effective treatments for Medtronic, Merck, Novartis, NeuroPhage Pharmaceuticals, Teva, and
UCB; has received honoraria from Medtronic, Teva, UCB, and Abbvie;
dyssynergic defecation. has received grants from Brain Canada, Canadian Institutes of Health
The potential roles of H pylori infection and SIBO in Research, Edmond J Safra Philanthropic Foundation, the Michael J Fox
the development of gastrointestinal dysfunction in Foundation, the Ontario Brain Institute, National Parkinson
Parkinson’s disease might allow new treatment Foundation, Parkinson Society Canada, Tourette Syndrome Association,

www.thelancet.com/neurology Vol 14 June 2015 635


Review

and the W Garfield Weston Foundation; and has served as an expert 24 Eadie MJ, Tyrer JH. Alimentary disorder in Parkinsonism.
witness in cases related to the welding industry. RFP has received Austral Ann Med 1965; 14: 13–22.
honoraria from Teva, UCB, and US WorldMeds for lectures; from 25 Edwards LL, Pfeiffer RF, Quigley EM, Hofman R, Balluff M.
Pfizer, Chelsea, and Acadia for consulting; and from UCB for research Gastrointestinal symptoms in Parkinson’s disease. Mov Disord 1991;
grants and contracts. NPV declares no competing interests. 6: 151–56.
26 Edwards LL, Quigley EM, Harned RK, Hofman R, Pfeiffer RF.
References Characterization of swallowing and defecation in Parkinson’s
1 Pfeiffer RF. Gastrointestinal dysfunction in Parkinson’s disease. disease. Am J Gastroenterol 1994; 89: 15–25.
Lancet Neurol 2003; 2: 107–16.
27 Siddiqui MF, Rast S, Lynn MJ, Auchus AP, Pfeiffer RF. Autonomic
2 Beach TG, Adler CH, Sue LI, et al. Multi-organ distribution of dysfunction in Parkinson’s disease: a comprehensive symptom
phosphorylated alpha-synuclein histopathology in subjects with survey. Parkinsonism Relat Disord 2002; 8: 277–84.
Lewy body disorders. Acta Neuropathol 2010; 119: 689–702.
28 Verbaan D, Marinus J, Visser M, van Rooden SM, Stiggelbout AM,
3 Gold A, Turkalp ZT, Munoz DG. Enteric alpha-synuclein van Hilten JJ. Patient-reported autonomic symptoms in Parkinson
expression is increased in Parkinson’s disease but not Alzheimer’s disease. Neurology 2007; 69: 333–41.
disease. Movement Disord 2013; 28: 237–40.
29 Nicaretta DH, de Rosso AL, Maliska C, Costa MM. Scintigraphic
4 Bottner M, Zorenkov D, Hellwig I, et al. Expression pattern and analysis of the parotid glands in patients with sialorrhea and
localization of alpha-synuclein in the human enteric nervous Parkinson’s disease. Parkinsonism Relat Disord 2008; 14: 338–41.
system. Neurobiol Dis 2012; 48: 474–80.
30 Kim HJ, Park SY, Cho YJ, et al. Nonmotor symptoms in de novo
5 Braak H, Rub U, Gai WP, Del Tredici K. Idiopathic Parkinson’s Parkinson disease before and after dopaminergic treatment.
disease: possible routes by which vulnerable neuronal types may J Neurol Sci 2009; 287: 200–04.
be subject to neuroinvasion by an unknown pathogen.
31 Leibner J, Ramjit A, Sedig L, et al. The impact of and the factors
J Neural Transm 2003; 110: 517–36.
associated with drooling in Parkinson’s disease.
6 Wakabayashi K, Takahashi H, Takeda S, Ohama E, Ikuta F. Parkinsonism Relat Disord 2010; 16: 475–77.
Parkinson’s disease: the presence of Lewy bodies in Auerbach’s
32 Yu B, Xiao ZY, Li JZ, Yuan J, Liu YM. Study of an integrated
and Meissner’s plexuses. Acta Neuropathol 1988; 76: 217–21.
non-motor symptoms questionnaire for Parkinson’s disease.
7 Gray MT, Munoz DG, Gray DA, Schlossmacher MG, Woulfe JM. Chin Med J 2010; 123: 1436–40.
Alpha-synuclein in the appendiceal mucosa of neurologically
33 Muller B, Larsen JP, Wentzel-Larsen T, Skeie GO, Tysnes OB.
intact subjects. Mov Disord 2014; 29: 991–98.
Autonomic and sensory symptoms and signs in incident,
8 Beach TG, Adler CH, Dugger BN, et al. Submandibular gland untreated Parkinson’s disease: frequent but mild. Mov Disord
biopsy for the diagnosis of Parkinson disease. 2011; 26: 65–72.
J Neuropathol Exp Neurol 2013; 72: 130–36.
34 Damian A, Adler CH, Hentz JG, et al. Autonomic function, as
9 Adler CH, Dugger BN, Hinni ML, et al. Submandibular gland self-reported on the SCOPA-autonomic questionnaire, is normal in
needle biopsy for the diagnosis of Parkinson disease. Neurology essential tremor but not in Parkinson’s disease.
2014; 82: 858–64. Parkinsonism Relat Disord 2012; 18: 1089–93.
10 Lebouvier T, Neunlist M, Bruley des Varannes S, et al. Colonic 35 Picillo M, Amboni M, Erro R, et al. Gender differences in
biopsies to assess the neuropathology of Parkinson’s disease and non-motor symptoms in early, drug naive Parkinson’s disease.
its relationship with symptoms. PloS One 2010; 5: e12728. J Neurol 2013; 260: 2849–55.
11 Visanji NP, Marras C, Kern DS, et al. Colonic mucosal 36 Shah M, Deeb J, Fernando M, et al. Abnormality of taste and smell
alpha-synuclein lacks specificity as a biomarker for Parkinson in Parkinson’s disease. Parkinsonism Relat Disord 2009; 15: 232–37.
disease. Neurology 2015; 84: 609–16.
37 Deeb J, Shah M, Muhammed N, et al. A basic smell test is as
12 Shannon KM, Keshavarzian A, Mutlu E, et al. Alpha-synuclein in sensitive as a dopamine transporter scan: comparison of olfaction,
colonic submucosa in early untreated Parkinson’s disease. taste and DaTSCAN in the diagnosis of Parkinson’s disease.
Movement Disord 2012; 27: 709–15. Q JM 2010; 103: 941–52.
13 Visanji NP, Marras C, Hazrati LN, Liu LW, Lang AE. Alimentary, 38 Kashihara K, Hanaoka A, Imamura T. Frequency and characteristics
my dear Watson? The challenges of enteric α-synuclein as a of taste impairment in patients with Parkinson’s disease: results of
Parkinson’s disease biomarker. Mov Disord 2014; 29: 444–50. a clinical interview. Intern Med 2011; 50: 2311–15.
14 Visanji NP, Brooks PL, Hazrati LN, Lang AE. The prion hypothesis 39 Pont-Sunyer C, Hotter A, Gaig C, et al. The Onset of Nonmotor
in Parkinson’s disease: Braak to the future. Symptoms in Parkinson’s disease (The ONSET PD Study).
Acta Neuropathol Commun 2013; 1: 2. Mov Disord 2014; 30: 229–237.
15 Gray MT, Munoz DG, Gray DA, Schlossmacher MG, Woulfe JM. 40 Clarke CE, Gullaksen E, Macdonald S, Lowe F. Referral criteria for
α-synuclein in the appendiceal mucosa of neurologically intact speech and language therapy assessment of dysphagia caused by
subjects. Mov Disord 2014; 29: 991–83. idiopathic Parkinson’s disease. Acta Neurol Scand 1998; 97: 27–35.
16 Cersosimo M, Benarroch E. Pathological correlates of gastrointestinal 41 Singer C, Weiner WJ, Sanchez-Ramos JR. Autonomic dysfunction
dysfunction in Parkinson’s disease. Neurobiol Dis 2012; 46: 559–64. in men with Parkinson’s disease. Eur Neurol 1992; 32: 134–40.
17 Pfeiffer RF. Parkinson’s disease and the gut: ‘the wheel is come full 42 Wang SJ, Fuh JL, Shan DE, et al. Sympathetic skin response and
circle’. J Parkinsons Dis 2014; 4: 577–78. R-R interval variation in Parkinson’s disease. Mov Disord 1993;
18 Noorian AR, Rha J, Annerino DM, Bernhard D, Taylor GM, 8: 151–57.
Greene JG. Alpha-synuclein transgenic mice display age-related 43 Gonera EG, van’t Hof M, Berger HJ, van Weel C, Horstink MW.
slowing of gastrointestinal motility associated with transgene Symptoms and duration of the prodromal phase in Parkinson’s
expression in the vagal system. Neurobiol Dis 2012; 48: 9–19. disease. Mov Disord 1997; 12: 871–76.
19 Muller T, Palluch R, Jackowski J. Caries and periodontal disease in 44 Abbott RD, Petrovitch H, White LR, et al. Frequency of bowel
patients with Parkinson’s disease. Spec Care Dentist 2011; movements and the future risk of Parkinson’s disease. Neurology
31: 178–81. 2001; 57: 456–62.
20 Bakke M, Larsen SL, Lautrup C, Karlsborg M. Orofacial function 45 Sakakibara R, Odaka T, Uchiyama T, et al. Colonic transit time and
and oral health in patients with Parkinson’s disease. Eur J Oral Sci rectoanal videomanometry in Parkinson’s disease.
2011; 119: 27–32. J Neurol Neurosurg Psychiatry 2003; 74: 268–72.
21 Hanaoka A, Kashihara K. Increased frequencies of caries, 46 Chaudhuri KR, Martinez-Martin P, Schapira AH, et al.
periodontal disease and tooth loss in patients with Parkinson’s International multicenter pilot study of the first comprehensive
disease. J Clin Neurosci 2009; 16: 1279–82. self-completed nonmotor symptoms questionnaire for Parkinson’s
22 Srivanitchapoom P, Pandey S, Hallett M. Drooling in Parkinson’s disease: the NMSQuest study. Mov Disord 2006; 21: 916–23.
disease: a review. Parkinsonism Relat Disord 2014; 20: 1109–118. 47 Kaye J, Gage H, Kimber A, Storey L, Trend P. Excess burden of
23 Nobrega AC, Rodrigues B, Melo A. Is silent aspiration a risk factor constipation in Parkinson’s disease: a pilot study. Mov Disord 2006;
for respiratory infection in Parkinson’s disease patients? 21: 1270–73.
Parkinsonism Relat Disord 2008; 14: 646–48.

636 www.thelancet.com/neurology Vol 14 June 2015


Review

48 Krogh K, Ostergaard K, Sabroe S, Laurberg S. Clinical aspects of 72 Ceravolo R, Cossu G, Bandettini di Poggio M, et al. Neuropathy and
bowel symptoms in Parkinson’s disease. Acta Neurol Scand 2008; levodopa in Parkinson’s disease: evidence from a multicenter study.
117: 60–64. Mov Disord 2013; 28: 1391–97.
49 Savica R, Carlin JM, Grossardt BR, et al. Medical records 73 Sauleau P, Le Jeune F, Drapier S, et al. Weight gain following
documentation of constipation preceding Parkinson disease: a subthalamic nucleus deep brain stimulation: a PET study.
case-control study. Neurology 2009; 73: 1752–58. Mov Disord 2014; 29: 1781–87.
50 Ramjit AL, Sedig L, Leibner J, et al. The relationship between 74 Heetun ZS, Quigley EM. Gastroparesis and Parkinson’s disease:
anosmia, constipation, and orthostasis and Parkinson’s disease a systematic review. Parkinsonism Relat Disord 2012; 18: 433–40.
duration: results of a pilot study. Int J Neurosci 2010; 120: 67–70. 75 Goetze O, Nikodem AB, Wiezcorek J, et al. Predictors of gastric
51 Gaenslen A, Swid I, Liepelt-Scarfone I, Godau J, Berg D. The emptying in Parkinson’s disease. Neurogastroenterol Motil 2006;
patients’ perception of prodromal symptoms before the initial 18: 369–75.
diagnosis of Parkinson’s disease. Mov Disord 2011; 26: 653–58. 76 Parkman HP, Hasler WL, Fisher RS. American Gastroenterological
52 Szewczyk-Krolikowski K, Tomlinson P, Nithi K, et al. The influence Association technical review on the diagnosis and treatment of
of age and gender on motor and non-motor features of early gastroparesis. Gastroenterology 2004; 127: 1592–622.
Parkinson’s disease: initial findings from the Oxford Parkinson 77 Zheng LF, Song J, Fan RF, et al. The role of the vagal pathway and
Disease Center (OPDC) discovery cohort. Parkinsonism Relat Disord gastric dopamine in the gastroparesis of rats after a
2014; 20: 99–105. 6-hydroxydopamine microinjection in the substantia nigra.
53 Schrag A, Horsfall L, Walters K, Noyce A, Petersen I. Prediagnostic Acta physiol (Oxf) 2014; 211: 434–46.
presentations of Parkinson’s disease in primary care: a case-control 78 Braak H, de Vos RA, Bohl JR, Del Tredici K. Gastric alpha-synuclein
study. Lancet Neurol 2015; 14: 57–64. immunoreactive inclusions in Meissner’s and Auerbach’s plexuses
54 Rana AQ, Yousuf MS, Awan N, Fattah A. Impact of progression of in cases staged for Parkinson’s disease-related brain pathology.
Parkinson’s disease on drooling in various ethnic groups. Neurosci Lett 2006; 396: 67–72.
Eur Neurol 2012; 67: 312–14. 79 Cheung CK, Wu JC. Role of ghrelin in the pathophysiology of
55 Kalf JG, Munneke M, van den Engel-Hoek L, et al. Pathophysiology gastrointestinal disease. Gut Liver 2013; 7: 505–12.
of diurnal drooling in Parkinson’s disease. Mov Disord 2011; 80 Nielsen HH, Qiu J, Friis S, Wermuth L, Ritz B. Treatment for
26: 1670–76. Helicobacter pylori infection and risk of Parkinson’s disease in
56 Kalf JG, Smit AM, Bloem BR, Zwarts MJ, Munneke M. Impact of Denmark. Eur J Neurol 2012; 19: 864–69.
drooling in Parkinson’s disease. J Neurol 2007; 254: 1227–32. 81 Dobbs RJ, Dobbs SM, Weller C, et al. Role of chronic infection and
57 Cersosimo MG, Raina GB, Calandra CR, et al. Dry mouth: an inflammation in the gastrointestinal tract in the etiology and
overlooked autonomic symptom of Parkinson’s disease. pathogenesis of idiopathic parkinsonism. Part 1: eradication of
J Parkinsons Dis 2011; 1: 169–73. Helicobacter in the cachexia of idiopathic parkinsonism. Helicobacter
58 Cecchini MP, Osculati F, Ottaviani S, Boschi F, Fasano A, 2005; 10: 267–75.
Tinazzi M. Taste performance in Parkinson’s disease. 82 Fasano A, Bove F, Gabrielli M, et al. The role of small intestinal
J Neural Transm 2014; 121: 119–22. bacterial overgrowth in Parkinson’s disease. Mov Disord 2013;
59 Coon EA, Laughlin RS. Burning mouth syndrome in Parkinson’s 28: 1241–49.
disease: dopamine as cure or cause? J Headache Pain 2012; 83 Narozanska E, Bialecka M, Adamiak-Giera U, et al.
13: 255–57. Pharmacokinetics of levodopa in patients with Parkinson disease
60 Kalf JG, de Swart BJ, Bloem BR, Munneke M. Prevalence of and motor fluctuations depending on the presence of Helicobacter
oropharyngeal dysphagia in Parkinson’s disease: a meta-analysis. pylori infection. Clin Neuropharmacol 2014; 37: 96–99.
Parkinsonism Relat Disord 2012; 18: 311–15. 84 Tan AH, Mahadeva S, Thalha AM, et al. Small intestinal bacterial
61 Lin CW, Chang YC, Chen WS, Chang K, Chang HY, Wang TG. overgrowth in Parkinson’s disease. Parkinsonism Relat Disord 2014;
Prolonged swallowing time in dysphagic Parkinsonism patients 20: 535–40.
with aspiration pneumonia. Archs Phys Med Rehabil 2012; 85 Isomoto H, Ueno H, Saenko VA, et al. Impact of Helicobacter pylori
93: 2080–84. infection on gastric and plasma ghrelin dynamics in humans.
62 Pfeiffer RF. Gastrointestinal dysfunction in Parkinson’s disease. Am J Gastroenterol 2005; 100: 1711–20.
Parkinsonism Relat Disord 2011; 17: 10–5. 86 Gabrielli M, Bonazzi P, Scarpellini E, et al. Prevalence of small
63 Noyce AJ, Silveira-Moriyama L, Gilpin P, Ling H, Howard R, intestinal bacterial overgrowth in Parkinson’s disease. Mov Disord
Lees AJ. Severe dysphagia as a presentation of Parkinson’s disease. 2011; 26: 889–92.
Mov Disord 2012; 27: 457–58. 87 Lee WY, Yoon WT, Shin HY, Jeon SH, Rhee PL. Helicobacter pylori
64 Kikuchi A, Baba T, Hasegawa T, et al. Hypometabolism in the infection and motor fluctuations in patients with Parkinson’s
supplementary and anterior cingulate cortices is related to disease. Mov Disord 2008; 23: 1696–700.
dysphagia in Parkinson’s disease: a cross-sectional and 3-year 88 Rahne KE, Tagesson C, Nyholm D. Motor fluctuations and
longitudinal cohort study. BMJ open 2013; 3: e002249. Helicobacter pylori in Parkinson’s disease. J Neurol 2013;
65 Mu L, Sobotka S, Chen J, et al. Altered pharyngeal muscles in 260: 2974–80.
Parkinson disease. J Neuropathol Exper Neur 2012; 71: 520–30. 89 Pierantozzi M, Pietroiusti A, Brusa L, et al. Helicobacter pylori
66 Sheard JM, Ash S, Silburn PA, Kerr GK. Prevalence of malnutrition eradication andL-dopa absorption in patients with PD and motor
in Parkinson’s disease: a systematic review. Nutr Rev 2011; fluctuations. Neurology 2006; 66: 1824–29.
69: 520–32. 90 Grace E, Shaw C, Whelan K, Andreyev HJ. Review article: small
67 Fereshtehnejad SM, Ghazi L, Shafieesabet M, Shahidi GA, intestinal bacterial overgrowth—prevalence, clinical features,
Delbari A, Lokk J. Motor, psychiatric and fatigue features associated current and developing diagnostic tests, and treatment.
with nutritional status and its effects on quality of life in Aliment Pharmacol Ther 2013; 38: 674–88.
Parkinson’s disease patients. PloS One 2014; 9: e91153. 91 Quigley EM, Quera R. Small intestinal bacterial overgrowth: roles
68 Barichella M, Cereda E, Pezzoli G. Major nutritional issues in the of antibiotics, prebiotics, and probiotics. Gastroenterology 2006;
management of Parkinson’s disease. Mov Disord 2009; 130: S78–90.
24: 1881–92. 92 Devos D, Lebouvier T, Lardeux B, et al. Colonic inflammation in
69 Kistner A, Lhommee E, Krack P. Mechanisms of body weight Parkinson’s disease. Neurobiol Dis 2013; 50: 42–48.
fluctuations in Parkinson’s disease. Front Neurol 2014; 5: 84. 93 Lomax AE, Linden DR, Mawe GM, Sharkey KA. Effects of
70 Laudisio A, Vetrano DL, Meloni E, et al. Dopaminergic agents and gastrointestinal inflammation on enteroendocrine cells and
nutritional status in Parkinson’s disease. Mov Disord 2014; enteric neural reflex circuits. Auton Neurosci 2006;
29: 1543–47. 126–127: 250–57.
71 Adams F, Boschmann M, Lobsien E, et al. Influences of levodopa on 94 Scheperjans F, Aho V, Pereira PA, et al. Gut microbiota are related
adipose tissue and skeletal muscle metabolism in patients with to Parkinson’s disease and clinical phenotype. Mov Disord 2014;
idiopathic Parkinson’s disease. Eur J Clin Pharmacol 2008; 64: 863–70. 30: 350–58.

www.thelancet.com/neurology Vol 14 June 2015 637


Review

95 Forsyth CB, Shannon KM, Kordower JH, et al. Increased 118 Kushnir M, Eilam A, Heldman E. Modafinil reduces drooling in
intestinal permeability correlates with sigmoid mucosa Parkinson’s disease. Move Disord 2006; 21 (suppl 13): S598–99.
alpha-synuclein staining and endotoxin exposure markers in early 119 Jost WH. Treatment of drooling in Parkinson’s disease with
Parkinson’s disease. PloS One 2011; 6: e28032. botulinum toxin. Mov Disord 1999; 14: 1057.
96 Lin C, Lin J, Liu Y, Chang C, Wu R. Risk of Parkinson’s disease 120 Pal PK, Calne DB, Calne S, Tsui JK. Botulinum toxin A as treatment
following severe constipation: a nationwide population-based for drooling saliva in PD. Neurology 2000; 54: 244–47.
cohort study. Parkinsonism Relat Disord 2014; 20: 1371–75. 121 Su CS, Lan MY, Liu JS, et al. Botulinum toxin type A treatment for
97 Postuma RB, Gagnon JF, Pelletier A, Montplaisir J. Prodromal Parkinsonian patients with moderate to severe sialorrhea.
autonomic symptoms and signs in Parkinson’s disease and Acta Neurologica Taiwanica 2006; 15: 170–76.
dementia with Lewy bodies. Mov Disord 2013; 28: 597–604. 122 Santamato A, Ianieri G, Ranieri M, et al. Botulinum toxin type A in
98 Wedel T, Spiegler J, Soellner S, et al. Enteric nerves and the treatment of sialorrhea in Parkinson’s disease. J Am Geriatr Soc
interstitial cells of Cajal are altered in patients with slow-transit 2008; 56: 765–67.
constipation and megacolon. Gastroenterology 2002; 123: 1459–67. 123 Friedman A, Potulska A. Quantitative assessment of parkinsonian
99 Annerino DM, Arshad S, Taylor GM, Adler CH, Beach TG, sialorrhea and results of treatment with botulinum toxin.
Greene JG. Parkinson’s disease is not associated with Parkinsonism Relat Disord 2001; 7: 329–32.
gastrointestinal myenteric ganglion neuron loss. Acta Neuropathol 124 Dogu O, Apaydin D, Sevim S, Talas DU, Aral M. Ultrasound-guided
2012; 124: 665–80. versus ‘blind’ intraparotid injections of botulinum toxin-A for the
100 Lebouvier T, Chaumette T, Damier P, et al. Pathological lesions in treatment of sialorrhoea in patients with Parkinson’s disease.
colonic biopsies during Parkinson’s disease. Gut 2008; Clin Neurol Neurosurg 2004; 106: 93–96.
57: 1741–43. 125 Lagalla G, Millevolte M, Capecci M, Provinciali L, Ceravolo MG.
101 Jost WH. Gastrointestinal dysfunction in Parkinson’s Disease. Botulinum toxin type A for drooling in Parkinson’s disease: a
J Neurol Sci 2010; 289: 69–73. double-blind, randomized, placebo-controlled study. Mov Disord
102 Liu L. Chronic constipation: current treatment options. 2006; 21: 704–07.
Can J Gastroenterol 2011; 25: 22B–28B. 126 Guidubaldi A, Fasano A, Ialongo T, et al. Botulinum toxin A versus
103 Malfertheiner P, Megraud F, O’Morain CA, et al. Management of B in sialorrhea: a prospective, randomized, double-blind, crossover
Helicobacter pylori infection—the Maastricht IV/Florence pilot study in patients with amyotrophic lateral sclerosis or
Consensus Report. Gut 2012; 61: 646–64. Parkinson’s disease. Mov Disord 2011; 26: 313–19.
104 Peedikayil M, Alsohaibani F, Alkhenizan A. Levofloxacin-based 127 Nobrega AC, Rodrigues B, Torres AC, Enzo A, Melo A. Does
first-line therapy versus standard first-line therapy for botulinum toxin decrease frequency and severity of sialorrhea in
Helicobacter pylori eradication: meta-analysis of randomized Parkinson’s disease? J Neurol Sci 2007; 253: 85–87.
controlled trials. PloS One 2014; 9: e85620. 128 Lipp A, Trottenberg T, Schink T, Kupsch A, Arnold G. A
105 Martin-Harris B, Jones B. The videofluorographic swallowing randomized trial of botulinum toxin A for treatment of drooling.
study. Phys MedRehabil Clin North Am 2008; 19: 769–85, viii. Neurology 2003; 61: 1279–81.
106 Sung HY, Kim JS, Lee KS, et al. The prevalence and patterns of 129 Mancini F, Zangaglia R, Cristina S, et al. Double-blind, placebo-
pharyngoesophageal dysmotility in patients with early stage controlled study to evaluate the efficacy and safety of botulinum
Parkinson’s disease. Mov Disord 2010; 25: 2361–68. toxin type A in the treatment of drooling in parkinsonism.
107 van Hooren MR, Baijens LW, Voskuilen S, Oosterloo M, Mov Disord 2003; 18: 685–88.
Kremer B. Treatment effects for dysphagia in Parkinson’s 130 Racette BA, Good L, Sagitto S, Perlmutter JS. Botulinum toxin B
disease: a systematic review. Parkinsonism Relat Disord 2014; reduces sialorrhea in parkinsonism. Mov Disord 2003; 18: 1059–61.
20: 800–07. 131 Contarino MF, Pompili M, Tittoto P, et al. Botulinum toxin B
108 Camilleri M, Shin A. Novel and validated approaches for gastric ultrasound-guided injections for sialorrhea in amyotrophic lateral
emptying scintigraphy in patients with suspected gastroparesis. sclerosis and Parkinson’s disease. Parkinsonism Relat Disord 2007;
Dig Dis Sci 2013; 58: 1813–15. 13: 299–303.
109 Bharucha AE, Pemberton JH, Locke GR 3rd. American 132 Ondo WG, Hunter C, Moore W. A double-blind placebo-controlled
Gastroenterological Association technical review on constipation. trial of botulinum toxin B for sialorrhea in Parkinson’s disease.
Gastroenterology 2013; 144: 218–38. Neurology 2004; 62: 37–40.
110 Packer M, Nikitin V, Coward T, Davis DM, Fiske J. The potential 133 Lagalla G, Millevolte M, Capecci M, Provinciali L, Ceravolo MG.
benefits of dental implants on the oral health quality of life of Long-lasting benefits of botulinum toxin type B in Parkinson’s
people with Parkinson’s disease. Gerodontology 2009; 26: 11–18. disease-related drooling. J Neurol 2009; 256: 563–67.
111 South AR, Somers SM, Jog MS. Gum chewing improves swallow 134 Chinnapongse R, Gullo K, Nemeth P, Zhang Y, Griggs L. Safety and
frequency and latency in Parkinson patients: a preliminary study. efficacy of botulinum toxin type B for treatment of sialorrhea in
Neurology 2010; 74: 1198–202. Parkinson’s disease: a prospective double-blind trial. Mov Disord
112 Arbouw ME, Movig KL, Koopmann M, et al. Glycopyrrolate for 2012; 27: 219–26.
sialorrhea in Parkinson disease: a randomized, double-blind, 135 Menezes C, Melo A. Does levodopa improve swallowing dysfunction
crossover trial. Neurology 2010; 74: 1203–07. in Parkinson’s disease patients? J Clin Pharm Ther 2009; 34: 673–6.
113 Seppi K, Weintraub D, Coelho M, et al. The movement disorder 136 Sutton JP. Dysphagia in Parkinson’s disease is responsive to
society evidence-based medicine review update: treatments for the levodopa. Parkinsonism Relat Disord 2013; 19: 282–84.
non-motor symptoms of Parkinson’s disease. Mov Disord 2011; 137 Troche MS, Brandimore AE, Foote KD, Okun MS. Swallowing and
26: S42–80. deep brain stimulation in Parkinson’s disease: a systematic review.
114 Hyson HC, Johnson AM, Jog MS. Sublingual atropine for Parkinsonism Relat Disord 2013; 19: 783–88.
sialorrhea secondary to parkinsonism: a pilot study. Mov Disord 138 Sheard JM, Ash S, Mellick GD, Silburn PA, Kerr GK. Markers of
2002; 17: 1318–20. disease severity are associated with malnutrition in Parkinson’s
115 Thomsen TR, Galpern WR, Asante A, Arenovich T, Fox SH. disease. PloS One 2013; 8: e57986.
Ipratropium bromide spray as treatment for sialorrhea in 139 Camilleri M, Parkman H, Shafi M, Abell T, Gerson L. Clinical
Parkinson’s disease. Mov Disord 2007; 22: 2268–73. guideline: management of gastroparesis. Am J Gastroenterol 2013;
116 Lloret SP, Nano G, Carrosella A, Gamzu E, Merello M. A double- 108: 18–37.
blind, placebo-controlled, randomized, crossover pilot study of the 140 Larson JM, Tavakkoli A, Drane WE, Toskes PP, Moshiree B.
safety and efficacy of multiple doses of intra-oral tropicamide films Advantages of azithromycin over erythromycin in improving the
for the short-term relief of sialorrhea symptoms in Parkinson’s gastric emptying half-time in adult patients with gastroparesis.
disease patients. J Neurol Sci 2011; 310: 248–50. JNeurogastroenterol Motil 2010; 16: 407–13.
117 Serrano-Duenas M. Treatment of sialorrhea in Parkinson’s disease 141 Doi H, Sakakibara R, Sato M, et al. Nizatidine ameliorates
patients with clonidine. Double-blind, comparative study with gastroparesis in Parkinson’s disease: a pilot study. Mov Disord 2014;
placebo. Neurologia 2003; 18: 2–6 (in Spanish). 29: 562–66.

638 www.thelancet.com/neurology Vol 14 June 2015


Review

142 McCallum RW, Lembo A, Esfandyari T, et al. Phase 2b, randomized, 160 Pfeiffer RF, Gutmann L, Hull KL Jr, et al. Continued efficacy and
double-blind 12-week studies of TZP-102, a ghrelin receptor agonist safety of subcutaneous apomorphine in patients with advanced
for diabetic gastroparesis. Neurogastroenterol Motil 2013; 25: e705–17. Parkinson’s disease. Parkinsonism Relat Disord 2007; 13: 93–100.
143 Bai Y, Xu MJ, Yang X, et al. A systematic review on intrapyloric 161 Schuepbach WM, Rau J, Knudsen K, et al. Neurostimulation for
botulinum toxin injection for gastroparesis. Digestion 2010; Parkinson’s disease with early motor complications. N Engl J Med
81: 27–34. 2013; 368: 610–22.
144 Arai E, Arai M, Uchiyama T, et al. Subthalamic deep brain 162 Olanow CW, Kieburtz K, Odin P, et al. Continuous intrajejunal
stimulation can improve gastric emptying in Parkinson’s disease. infusion of levodopa-carbidopa intestinal gel for patients with
Brain 2012; 135: 1478–85. advanced Parkinson’s disease: a randomised, controlled,
145 Jayanthi NV, Dexter SP, Sarela AI. Gastric electrical stimulation for double-blind, double-dummy study. Lancet Neurol 2014; 13: 141–49.
treatment of clinically severe gastroparesis. J Minim Access Surg 163 Fasano A, Bove F, Gabrielli M, et al. Liquid melevodopa versus
2013; 9: 163–67. standard levodopa in patients with Parkinson disease and small
146 Gatto M, Fernandez Pardal M, Melero M, Zurru C, Scorticati C, intestinal bacterial overgrowth. Clin Neuropharmacol 2014;
Micheli F. L-dopa malabsorption in a parkinsonian patient with 37: 91–95.
Strongyloides stercoralis duodenitis. Clin Neuropharmacol 1994; 164 Stocchi F, Fabbri L, Vecsei L, Krygowska-Wajs A, Monici Preti PA,
17: 96–98. Ruggieri SA. Clinical efficacy of a single afternoon dose of effervescent
147 Yuan Y, Leontiadis G. Levofloxacin-based three-antibiotic regimen levodopa-carbidopa preparation (CHF 1512) in fluctuating Parkinson
for H. pylori eradication. Am J Gastroenterol 2012; 107: 1106–07. disease. Clinical Neuropharmacol 2007; 30: 18–24.
148 Stahlmann R, Lode H. Safety considerations of fluoroquinolones in 165 Fasano A, Ricciardi L, Lena F, Bentivoglio AR, Modugno N.
the elderly: an update. Drugs Aging 2010; 27: 193–209. Intrajejunal levodopa infusion in advanced Parkinson’s disease:
149 Lauritano EC, Gabrielli M, Scarpellini E, et al. Small intestinal long-term effects on motor and non-motor symptoms and impact
bacterial overgrowth recurrence after antibiotic therapy. on patient’s and caregiver’s quality of life.
AmJ Gastroenterol 2008; 103: 2031–35. Eur Rev Med Pharmacol Sci 2012; 16: 79–89.
150 Eichhorn T, Oertel W. Macrogol 3350/electrolyte improves 166 Frankel JP, Kempster PA, Bovingdon M, Webster R, Lees AJ,
constipation in Parkinson’s disease and multiple system atrophy. Stern GM. The effects of oral protein on the absorption of
Mov Disord 2001; 16: 1176–77. intraduodenal levodopa and motor performance.
J Neurol Neurosurg Psychiatry 1989; 52: 1063–67.
151 Ondo W, Kenney C, Sullivan K, et al. Placebo-controlled trial of
lubiprostone for constipation associated with Parkinson disease. 167 Ondo WG, Hunter C, Ferrara JM, Mostile G. Apomorphine
Neurology 2012; 78: 1650–54. injections: predictors of initial common adverse events and long
term tolerability. Parkinsonism Relat Disord 2012; 18: 619–22.
152 Lembo A, Johanson J, Parkman H, Rao S, Miner PJ, Ueno R.
Long-term safety and effectiveness of lubiprostone, a chloride 168 Antonini A, Isaias IU, Rodolfi G, et al. A 5-year prospective
channel (ClC-2) activator, in patients with chronic idiopathic assessment of advanced Parkinson disease patients treated with
constipation. Dig Dis Sci 2011; 56: 2639–45. subcutaneous apomorphine infusion or deep brain stimulation.
J Neurol 2011; 258: 579–85.
153 Sullivan K, Staffetti J, Hauser R, Dunne P, Zesiewicz T. Tegaserod
(Zelnorm) for the treatment of constipation in Parkinson’s disease. 169 Garcia-Ruiz PJ, Martinez Castrillo JC, Alonso-Canovas A, et al.
Mov Disord 2006; 21: 115–16. Impulse control disorder in patients with Parkinson’s disease under
dopamine agonist therapy: a multicentre study.
154 Lembo A, Schneier H, Shiff S, et al. Two randomized trials of
J Neurol Neurosurg Psychiatry 2014; 85: 840–44.
linaclotide for chronic constipation. N Engl J Med 2011; 365: 527–36.
170 Fasano A, Daniele A, Albanese A. Treatment of motor and
155 Albanese A, Maria G, Bentivoglio A, Brisinda G, Cassetta E,
non-motor features of Parkinson’s disease with deep brain
Tonali P. Severe constipation in Parkinson’s disease relieved by
stimulation. Lancet Neurol 2012; 11: 429–42.
botulinum toxin. Mov Disord 1997; 12: 764–66.
171 Iyer SS, Morgan JC, Sethi KD. Absorption of orally disintegrating
156 Ondo WG, Shinawi L, Moore S. Comparison of orally dissolving
carbidopa-levodopa requires intact small bowel function. Neurology
carbidopa/levodopa (Parcopa) to conventional oral carbidopa/
2005; 65: 1507.
levodopa: A single-dose, double-blind, double-dummy, placebo-
controlled, crossover trial. Mov Disord 2010; 25: 2724–27. 172 Camilleri M, Acosta A. Emerging treatments in
Neurogastroenterology: relamorelin: a novel gastrocolokinetic
157 Stocchi F, Zappia M, Dall’Armi V, et al. Melevodopa/carbidopa
synthetic ghrelin agonist. Neurogastroenterol Motil 2015; 27: 324–32.
effervescent formulation in the treatment of motor fluctuations in
advanced Parkinson’s disease. Mov Disord 2010; 25: 1881–87. 173 Aro P, Storskrubb T, Ronkainen J, et al. Peptic ulcer disease in a
general adult population: the Kalixanda study: a random
158 Jansson Y, Eriksson B, Johnels B. Dispersible levodopa has a fast
population-based study. Am J Epidemiol 2006; 163: 1025–34.
and more reproducible onset of action than the conventional
preparation in Parkinson’s disease. A study with optoelectronic
movement analysis. Parkinsonism Relat Disord 1998; 4: 201–06.
159 Giladi N, Boroojerdi B, Korczyn AD, et al. Rotigotine transdermal
patch in early Parkinson’s disease: a randomized, double-blind,
controlled study versus placebo and ropinirole. Mov Disord 2007;
22: 2398–404.

www.thelancet.com/neurology Vol 14 June 2015 639

You might also like