Next Article in Journal
Targeting Cancer Stem Cells to Overcome Chemoresistance
Next Article in Special Issue
Isodon rugosus (Wall. ex Benth.) Codd In Vitro Cultures: Establishment, Phytochemical Characterization and In Vitro Antioxidant and Anti-Aging Activities
Previous Article in Journal
Lymphatic Vascular Structures: A New Aspect in Proliferative Diabetic Retinopathy
Previous Article in Special Issue
Melanogenesis Inhibitors from the Rhizoma of Ligusticum Sinense in B16-F10 Melanoma Cells In Vitro and Zebrafish In Vivo
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

DA-9701 (Motilitone): A Multi-Targeting Botanical Drug for the Treatment of Functional Dyspepsia

1
Department of Microbiology, College of Medicine, Gachon University, Incheon 21999, Korea
2
Department of Health Science and Technology, GAIHST, Gachon University, Incheon 21936, Korea
3
Research Center & Phytotherapeutics Group, Viromed, Co. Ltd., Seoul 08826, Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2018, 19(12), 4035; https://doi.org/10.3390/ijms19124035
Submission received: 9 November 2018 / Revised: 7 December 2018 / Accepted: 7 December 2018 / Published: 13 December 2018
(This article belongs to the Special Issue Health Promoting Effects of Phytochemicals)

Abstract

:
Functional dyspepsia (FD) is the most common functional gastrointestinal disorder (FGID). FD is characterized by bothersome symptoms such as postprandial fullness, early satiety, and epigastric pain or burning sensations in the upper abdomen. The complexity and heterogeneity of FD pathophysiology, which involves multiple mechanisms, make both treatment and new drug development for FD difficult. Current medicines for FD targeting a single pathway have failed to show satisfactory efficacy and safety. On the other hand, multicomponent herbal medicines that act on multiple targets may be a promising alternative treatment for FD. DA-9701 (Motilitone), a botanical drug consisting of Corydalis Tuber and Pharbitidis Semen, has been prescribed for FD since it was launched in Korea in 2011. It has multiple mechanisms of action such as prokinetic effects, fundus relaxation, and visceral analgesia, which are mediated by dopamine D2 and several serotonin receptors involved in gastrointestinal (GI) functions. In clinical studies, DA-9701 has been found to be beneficial for improvement of FD symptoms and GI functions in FD patients, while showing better safety compared to that associated with conventional medicines. In this review, we provide updated information on the pharmacological effects, safety, and clinical results of DA-9701 for the treatment of FGIDs.

1. Introduction

1.1. Functional Dyspepsia: Evolution of Its Definition and Criteria

Functional gastrointestinal disorders (FGIDs) are the most common functional gastroenterologic abnormalities associated with physiological and morphological disturbances, and are often accompanied by conditions including motility dysfunction, visceral hypersensitivity, altered mucosal and immune functions, altered gut microbiota, and altered central nervous system processing [1,2,3]. In particular, functional dyspepsia (FD) has been recognized as an unexplained discomfort in the upper abdomen for over 100 years, and its symptoms include excessive fullness after eating or the inability to finish a normal-sized meal and recurrent epigastric pain [3,4]. FD is associated with not only a markedly impaired quality of life and negative impact on the work place, but also a significant economic burden [5]. The global prevalence of FD has been reported as 21%, with the range being 10% to 30% among various studies and geographies [6]. The definition and diagnostic criteria of FD have evolved within the Rome process [7]. The Rome Foundation was established in the late 1980s, at a time when there was little understanding of the pathophysiology of FGIDs. In Rome II criteria [8], FD was defined as recurrent upper abdominal pain and discomfort for at least 12 weeks during the preceding year. The Rome II classification divided patients having a wide range of dyspepsia symptoms into four groups on the basis of major symptomatic patterns: Reflux-like, ulcer-like, dysmotility-like, or nonspecific FD. However, the Rome II categorization lacked reliability, which was attributable to overlapping symptoms of various FGIDs such as irritable bowel syndrome (IBS), gastroesophageal reflux disease (GERD), postprandial distress syndrome (PDS), and epigastric pain syndrome (EPS). Therefore, the Rome III committee subdivided FD into two distinct syndromes: PDS characterized by postprandial fullness and early satiety, and EPS accompanied by epigastric pain or burning [9,10]. The Rome IV consensus published in 2016 emphasized that FD should not be considered a single disorder. It retained the subclassification of FD into PDS and EPS but strengthened the notion that these symptoms are separate entities that could overlap; it also emphasized that the symptoms should be severe enough to be bothersome (i.e., a discomfort score of at least 2 on a scale of 1 to 5 in daily life) and occur more frequently than that in the normal population. PDS and EPS were defined as “bothersome early satiety or postprandial fullness for three or more days per week in 3 months with at least a 6-month history” and “bothersome epigastric pain or epigastric burning for 1 or more days per week in the past 3 months with at least a 6-month history”, respectively [3,11]. In addition, the Rome IV consensus accepted evidence that the symptoms of GERD and IBS are part of the spectrum of FD, and the brain-gut axis is an important factor in the etiology of functional gastrointestinal (GI) disorders. It further acknowledged the possibility that pathologic lesions are involved in FD development, even if FD is a functional disorder, based on the findings that duodenal inflammation and eosinophilia are associated with FD [12,13,14]. Helicobacter pylori-associated dyspepsia is classified separately from true FD in the Rome IV criteria; affected patients are defined as a subset of those with FD-like symptoms and H. pylori infection, with symptom resolution 1 year after successful eradication of H. pylori [11,15].

1.2. Pathophysiology of FD and Current Treatments

Undoubtedly, FD is a complex and heterogeneous disorder. Accumulating data indicate that multiple factors including environment factors (food and H. pylori infection), biological factors (duodenal inflammation, eosinophilia, and cytokines), physiological factors (acid, gastric emptying, and gastric accommodation), and psychological factors (visceral hypersensitivity, brain pain modulating circuits, and anxiety/depression) are involved in the pathophysiology of FD [7] (Figure 1). The multiple mechanisms involved in FD make successful treatment and the development of new drugs with satisfactory efficacy and safety difficult.
Historically, FD has been considered a motility disorder dominated by disturbances in gastric physiology including gastric emptying and relaxation. Although gastric emptying was not correlated with symptoms [9,10], delayed gastric emptying was reported, with an incidence of 20 to 50% among FD patients, and the overall gastric emptying of solids was 1.5 times slower in FD patients than in healthy subjects [16]. Usually, a prokinetic is administered as a first-line treatment for PDS patients [6]. Approximately 90% of the human body’s total serotonin (5-hydroxytryptamine, 5-HT) is located in enterochromaffin cells in the gut. Intestinal movement is regulated via 5-HT receptors, including 5-HT1, 5-HT2, 5-HT3, 5-HT4, and 5-HT7, which are activated by serotonin as their natural ligand. Serotonin receptors are drug targets for FGIDs [17,18,19,20]. After the withdrawal of cisapride, a 5-HT4 agonist and 5-HT3 antagonist, because of its cardiovascular side effects, several serotonergic drugs including tegaserod and mosapride were launched into the market. However, the efficacy and safety of these drugs are limited [21,22]. Mosapride failed to provide any benefit [23] and tegaserod was withdrawn due to cardiovascular side effects [24].
Itopride, a prokinetic agent that works as a dual dopamine D2 receptor antagonist and acetylcholinesterase inhibitor, was reported to have good efficacy in global patient assessments and to improve FD symptom scores in a meta-analysis [25], but it failed to demonstrate any efficacy in a clinical trial [26]. Domperidone (a dopamine D2 and D3 receptor antagonist) and metoclopramide, (a dopamine D1 and D2 receptor antagonist) were also launched; however, their side effects, which included ventricular arrhythmia and extrapyramidal movement disorder, respectively, inhibited their long-term use [27,28].
Gastric accommodation is relaxation of the fundus, the upper part of the stomach, in response to food ingestion, which is mediated by a vasovagal reflex generated via nitrergic nerve activation. Tack et al. reported impaired gastric accommodation in 40% of FD patients [10], which was associated with early satiety [10]. Drugs that relax the gastric fundus appear to improve FD symptoms, particularly PDS. Buspirone, an antidepressant having a partial agonistic effect on 5-HT1A, may enhance relaxation of the gastric fundus and body, and reduce the severity of symptoms including postprandial fullness, early satiety, and upper abdominal bloating [29]. Sumatriptan, a 5-HT1B/D agonist, was reported to alleviate FD symptoms by restoration of gastric accommodation with fundus-relaxing effects [30]. Recently, acotiamide, an M1/M2 muscarinic receptor blocker, was approved as a fundic relaxant for FD in Japan. This drug increases acetylcholine availability in gut nerve synapses [31], and has been reported to relax the gastric fundus and accelerate gastric emptying in humans [32,33].
Gastric hypersensitivity and/or brain-gut dysfunction has been associated with an impaired connection between gastric physiology and psychology in FD [34]. Compared to that of normal subjects, patients with FGIDs have been reported to be more sensitive to balloon distension. Many studies have confirmed intolerance to gastric distention in patients with FD [35,36]. Gastric hypersensitivity seems to be associated with postprandial epigastric pain, belching, and weight loss [37]. Approximately 34% of patients with FD had a lower threshold for the first perception of stimulus, discomfort, and pain during distention of the proximal stomach by barostat [37,38]. Visceral analgesics including alosetron, a 5-HT3 antagonist [39], were reported to reduce visceral hypersensitivity and to be beneficial in symptom relief compared to that of placebo in clinical studies [40]. However, side effects, including constipation and ischemic colitis, were a concern [41]. Fedotozine, an opioid κ-agonist [42,43], seemed to be a visceral analgesic; however, in clinical trials, the results of an overall physician assessment of this drug were not satisfactory [44]. For EPS patients, acid suppression is beneficial, and proton pump inhibitors (PPIs) are primarily used. H2 receptor antagonist therapy was reported to be superior to placebo control to reduce FD symptoms [45]. However, if first-line therapies fail, centrally acting drugs may be beneficial. Tricyclic antidepressants, including amitriptyline, appeared to provide a modest benefit for FD patients, especially for those with pain [46]. Interestingly, the tetracyclic antidepressant mirtazapine, which is an H1 histamine receptor blocker, 5-HT2C and 5-HT3 antagonist, and α2 adrenergic receptor blocker, seemed to be more beneficial than placebo in a preliminary randomized controlled trial of FD patients with weight loss [47,48], although it did not alter gastric function in healthy individuals [49]. Levosulpiride, an atypical antipsychotic drug and dopamine antagonist, may be efficacious in FD treatment [50]. However, more data including those from large placebo-controlled trials are needed [51].
Given the lack of medications with satisfactory efficacy and safety and the requirement of multiple drugs to achieve relief from various symptoms, there remains an unmet need for new treatments. Researchers have proposed that an agent capable of modulating multiple mechanisms will be more promising than an agent highly selective for a single mechanism and that a new treatment for FD should target many, if not all, pathophysiologies [52].
For centuries, herbal preparations have been traditionally used for a variety of GI disorders based on historical experiences and oriental medicine practices, but there has been a lack of scientific evidence and qualified clinical data. Herbal medicines, which generally contain multiple herbs, may be good options for FD treatment. The pathophysiology of FD is heterogeneous and multiple components of herbal medicines may enable simultaneous targeting of the multiple pathways involved in FD, enhancing efficacy compared to that of current chemical drugs targeting a single pathway [52]. DA-9701 (Motilitone) was launched as a new drug for FD in December 2011 in Korea; it is an herbal medicine with multi-acting mechanisms for FD treatment [53]. In this review, we provide updated information related to the multi-acting pharmacological effects, safety, and clinical data of DA-9701 and propose further research on it.

2. DA-9701

2.1. Herbal Composition

DA-9701 is a botanical drug for FD treatment in Korea, which is formulated with Pharbitidis Semen and Corydalis Tuber [53]; both constituent plants have been traditionally used for treating GI disorders [54,55,56]. Corydalis Tuber, the root of Corydalis yanhusuo W.T. Wang (Papaveraceae), is known to control gastric juice secretion and prevent gastric [56] and duodenal ulcers [57]. Extracts from Corydalis Tuber have been used as antispasmodic agents and analgesics [54] for abdominal pain because of its soothing and tranquilizing properties. Anti-inflammatory effects of the extract or its isolated compounds have been reported [58]. Pharbitidis Semen is the seed of Pharbitis nil Choisy (Convolvulaceae) and has been used as a traditional medicine for the treatment of abdominal pain [55] and as a strong purgative in Chinese medicine. The extract of Pharbitidis Semen is also used to stimulate and enhance intestinal peristalsis. Compared to that of a drug comprising a single synthetized chemical compound, a botanical drug requires more complicated quality control because of the properties of natural products. DA-9701 contains various components including corydaline (CD), dehydrocorydalin, chlorogenic acid, caffeic acid, coptisine, berberine, tetrahydroberberine (THB), palmatine, and tetrahydropalmatine (THP). For batch to batch control, CD ((13S, 13aR)-2,3,9,10-tetramethoxy-13-methyl-6,8,13,13a-tetrahydro-5H-isoquinolino[2,1-b]), the main constituent of Corydalis Tuber and CA (1S, 3R, 4R, 5R0-3-[(E)-3-(3,4-)prop-2-enoyl]oxy-1,4,5-trihydroxycyclohexane-1-carboxylic acid) from Pharbitidis Semen were selected as marker compounds for DA-9701 [59].

2.2. Pharmacology of DA-9701

The in vitro and in vivo pharmacological study results of DA-9701 are summarized in Table 1.

2.3. Effects of DA-9701 on GI Motility

The effects of DA-9701 on gastric emptying have been evaluated in not only normal animals but also models of delayed gastric emptying induced by apomorphine, cisplatin, opioids, and clonidine. [77,78,79]. The prokinetic effects of DA-9701 were comparable to those of conventional drugs such as cisapride (10 mg/kg) in normal animals and a delayed gastric emptying model at doses of 0.3–3 mg/kg [60] and in an in vitro study of interstitial cells of Cajal, indicating pacemaker activity of DA-9701 [61]. Further, DA-9701 administration accelerated gastric emptying, as shown by a 13C-octanoic acid breath test with repeated measurement in normal mice [62]. In rats sutured with a strain gauge force transducer, DA-9701 administration significantly restored clonidine-induced hypomotility of the gastric antrum in pre- and postprandial periods [63]. When laparotomy, atropine, or opioids were administered to animals to examine the effects of DA-9701 on delayed GI transit, DA-9701 enhanced GI transit in mice who had been subjected to laparotomy or atropine injection [60]. Further, in guinea pigs with opioid-induced bowel dysfunction, delayed GI transit was restored and fecal output was increased. In organ bath experiments using morphine-treated ileal muscle with reduced contractility, DA-9701 significantly increased the amplitude of contraction [64]. Additionally, DA-9701 administration to animals with abdominal surgery was able to ameliorate postoperative ileus (POI) by reducing delayed GI transit and improving defecation [65]. These effects might be mediated by decreased expression of corticotrophin releasing factor in the hypothalamus of DA-9701-pretreated animals with POI [66]. Diverse stresses usually induce dyspeptic symptoms [80], probably by alteration of gastric sensory and motor functions, and acute stressors are known to be associated with delayed gastric emptying in animal models and humans [81,82]. The administration of DA-9701 improved delayed gastric emptying induced by stress, including immobilization and subsequent immersion in a water bath, which was associated with inhibition of stress-induced increases in plasma levels of adrenocorticotropic hormone (ACTH) and ghrelin [67]. DA-9701 has a high affinity for multiple receptors related to GI function. It enhances gastric emptying and GI transit via dopamine D2 antagonism and 5-HT4 agonism [83]. The affinities of DA-9701 for the D2 and 5-HT4 receptors were 0.381 and 13.2 µg/mL, respectively [53]. THB and THP, two components of DA-9701, showed dopamine D2 receptor antagonistic effects, and their IC50 values against GTPγS binding to recombinant human dopamine D2S receptor in Chinese hamster ovary cells were 0.622 µM (211 ng/mL) and 1.32 µM (469 ng/mL), respectively (Dong-A Pharmaceutical Co., Ltd., Seoul, South Korea). There is some controversy about the coexistence of relaxation and contraction effects on the stomach. However, this can be explained by the fact that contractile receptors are dominant in the antrum region and relaxing receptors are dominant in the fundus region of the stomach [84].

2.4. Effects of DA-9701 on Fundic Relaxation

Currently, the barostat is the most commonly available method for measuring fundus relaxation in human patients and canine models [37,85]. To evaluate the fundus relaxation effect, two end points can be measured: Accommodation and compliance [86,87]. The gastric accommodation response enables proximal stomach relaxation to provide space for receiving foods without an increase in gastric pressure [88]. Meal-induced gastric accommodation is thought to be the most important motor index that can be studied currently. Gastric compliance, which is tested in the fasting state, is a measure of gastric tone in the resting state and seems to be related to the pain and discomfort perception threshold [87,89]. DA-9701 significantly increased gastric accommodation in Beagle dogs, shifting the pressure-volume curve toward higher volumes that were comparable to those associated with the control dogs [60]. This finding was reproduced in the same experimental system by CD, a component of DA-9701 [68]. In DA-9701-treated dogs, meal-induced gastric volumes significantly increased and persisted, a finding comparable to that achieved with sumatriptan [69]. DA-9701 may be effective in restoring restraint stress-induced food intake inhibition in rats, and this effect appears to be related to enhanced gastric accommodation by 5-HT1A agonism [70]. Further, THB isolated from DA-9701 alleviated impaired gastric compliance in the rat after stress induction and relaxed the proximal stomach via 5-TH1A agonism [71]. Agonism of 5-HT1A and 5-HT1B/D is known to mediate relaxation of the gastric fundus through activation of a nitric oxide pathway [87,90] and to decrease the visceromotor response to noxious colorectal distention via smooth muscle relaxation [89]. The affinity of DA-9701 to 5-HT1A was shown to be 6.87 μg/mL. In in vitro studies, DA-9701 inhibited 5-HT-induced contraction in feline esophageal smooth muscle cells by reducing the phosphorylation of myosin light chain kinase (MLC20) [72]. Using gastric fundus muscle strips triggered with electrical field stimulation, it was demonstrated that the effect of DA-9701 on rat gastric fundus relaxation is mainly mediated by nitrergic rather than purinergic pathways [73].

2.5. Effects of DA-9701 on Visceral Hypersensitivity

Visceral hypersensitivity is one of the leading targets of FD drug development. In a neonatal rat model of colon irritation with colorectal distention (CRD), administration of DA-9701 significantly decreased mean arterial pressure in a dose-dependent manner, indicating an increase in the pain threshold with CRD-induced visceral hypersensitivity [74]. Pain signals are transmitted from the peripheral regions to the spinal cord (dorsal root ganglion) where the information is processed for transfer to the central nervous system (CNS). In rats treated with DA-9701, phosphorylation of extracellular signal-regulated kinase (p-ERK), a pain-related factor of the pain transmission pathway, significantly decreased in response to CRD [76], suggesting modulation of visceral pain. Further, THP and CD, two components of DA-9701, were reported to have antinociceptive effects on visceral and somatic nociception in animals [75]. It is known that the adrenergic nervous system plays a certain role in modulating visceral nociceptive processing. Adrenergic α2 agonists, such as clonidine, have been shown to reduce pain perception during gastric and colonic distention [89] and to produce post-operative analgesia in humans [91]. This is probably mediated by modulation of spinal neurotransmitters at the levels of the dorsal horn, activation of descending inhibitory pathways, and/or emotional responses to visceral stimuli [92,93]. The affinity of DA-9701 to adrenergic α2 receptors was shown to be 4.81 μg/mL.

3. Safety of DA-9701

In a single-dose toxicity study, the LD50 of DA-9701 was over 2,000 mg/kg, and in a repeated-dose toxicity study for 26 weeks, the no-observed-adverse-effect level (NOAEL) was 150 mg/kg in rats. The NOAEL was 100 mg/kg in both 1-week and 13-week repeated-treatment studies in dogs. DA-9701 exhibited no genotoxicity. Due to the D2 antagonism of DA-9701, hyperprolactinemia was the major safety concern; however, the prolactin ED200 of DA-9701 was approximately 70-fold lower than that of itopride (3.78 vs. 270.1 mg/kg) in rats [53]. The pharmacokinetics and CNS distribution of THB and THP from DA-9701, both having dopamine D2 receptor binding affinities, were examined because D2 antagonists such as metoclopramide have a direct effect on the CNS by crossing the blood-brain barrier [59]. A tissue distribution study revealed that THB and THP were present at high concentrations in the stomach and small intestine compared to those in the plasma following administration of various oral doses of DA-9701, indicating considerable GI distribution. Increased concentrations of THB and THP, which cross the blood-brain barrier, were measured in the brain after repeated-dose administration of DA-9701. However, the brain concentration of DA-9701 was not expected to be sufficient to exert central dopamine D2 receptor antagonism following oral administration of effective doses in humans [59].

4. Clinical Studies of DA-9701

The therapeutic efficacy of DA-9701 has been investigated in several clinical trials according to modern guidelines and these clinical studies are summarized in Table 2.
The first clinical trial using DA-9701 was a multicenter, double-blind, randomized, and controlled trial with concealed allocation comparing the safety and efficacy of DA-9701 and itopride hydrochloride in Korea. Four hundred and sixty-four FD patients aged >20 years who were diagnosed with FD according to the Rome II criteria were randomized to the DA-9701 (30 mg t.i.d.) or itopride (50 mg t.i.d.) group. There was a 2-week medication-free run-in phase and a 4-week treatment period. Two primary efficacy end points, including the change in composite score from baseline of the eight dyspeptic symptoms and the overall treatment effect, were analyzed. Using the Nepean Dyspepsia Index (NDI) questionnaire, the impact on patient quality of life was assessed. DA-9701 significantly improved both FD symptoms and quality of life in patients with FD. The efficacy of DA-9701 was not inferior to that of itopride. Both drugs increased the NDI score of five domains without any differences between the groups. The safety profiles of both drugs were comparable. DA-9701 was well tolerated and did not show drug-related serious adverse effects [94].
To investigate the efficacy of DA-9701 for improving FD symptoms in comparison with that of pantoprazole, a PPI, and to evaluate the additive effect of DA-9701 over PPI treatment only, a multicenter, double-blind, randomized, parallel-comparative phase IV study was conducted. Three hundred and eighty-nine patients diagnosed with FD by using the Rome III criteria were allocated to three groups: 30 mg DA-9701 t.i.d., 40 mg pantoprazole q.d., and 30 mg DA-9701 t.i.d + 40 mg pantoprazole q.d. Patients in all treatment groups reported significant improvements in FD symptoms and dyspepsia-related quality of life (p < 0.001). The global symptomatic improvement was 60.5% in the DA-9701 group, 65.6% in the pantoprazole group, and 63.5% in the DA-9701 + pantoprazole group, as determined using a 5-point Likert scale at week 4; there were no significant intergroup differences [95].
In another study, 81 patients with minimal change esophagitis presenting with reflux or dyspeptic symptoms were enrolled and 42 and 39 patients were randomly assigned to receive either DA-9701 30 mg or placebo t.i.d., respectively. After 4 weeks, the NDI questionnaire-Korean version (NDI-K) symptom scores [96] were significantly reduced in the treatment (p < 0.001) and control groups (p < 0.001). However, changes in the symptom scores and quality of life scores did not differ between the two groups. The reflux symptom score significantly improved in the treatment group compared to that in the placebo group among patients aged 65 years or older (p = 0.035). Although the NDI-K symptom scores and quality of life scores improved after 4 weeks of treatment compared to baseline values in patients with minimal change esophagitis, DA-9701 did not improve the symptom scores or quality of life scores compared with those of the placebo [96].
While PD patients have impaired gastric function, which can cause altered responses to oral dopaminergic drugs, prokinetics with dopaminergic antagonism are commonly prescribed to prevent nausea and vomiting induced by anti-parkinsonian drugs. Therefore, to evaluate the clinical utility of DA-9701 in PD patients, the effects of DA-9701 on gastric motility were evaluated in PD patients by using magnetic resonance imaging (MRI). In a non-inferiority study with domperidone, 38 participants completed the 4-week treatment protocol. DA-9701 was not inferior to domperidone in terms of changes in the 2-h gastric emptying rate (GER) before and after treatment. However, a significant increase in the 2-h GER was observed only in the DA-9701 group (p < 0.05) without aggravation of PD symptoms [97].
A clinical study was conducted to evaluate the effects of DA-9701 on gastric accommodation and emptying after a meal in healthy volunteers by using 3-D gastric volume measurement with MRI. Forty healthy subjects randomly received DA-9701 or placebo. After 5 days of treatment, the subjects underwent gastric MRI (60 min before and 15, 30, 45, 60, 90, and 120 min after a liquid test meal). Whereas DA-9701 did not significantly affect gastric accommodation in healthy volunteers, pretreatment with DA-9701 increased postprandial proximal-to-distal total gastric volume ratio compared to that with placebo. Pretreatment with DA-9701 significantly increased gastric emptying compared to pretreatment with placebo [98].
In a recent prospective study conducted in 27 patients with functional constipation diagnosed based on the Rome III criteria, DA-9701 30 mg t.i.d. for 24 days was associated with a significantly reduced colonic transit time (CTT) (p = 0.001) and decreased segmental CTT (p < 0.001). In addition, all constipation-related subjective symptoms, including spontaneous bowel movement frequency, significantly improved compared to those before treatment, without any serious adverse effects [99].

5. Conclusion and Future Prospects

DA-9701, a natural product for FD treatment, is composed of multiple components that act on multiple targets involved in the pathophysiology of FD. DA-9701 has affinity for dopamine, serotonin, and adrenergic receptors. Therefore, it has multiple pharmacological actions through antagonistic effects on D2 and agonistic effects on 5-HT4, 5-HT1A, and 5-HT1B, which are associated with gastric emptying, relaxation, and hypersensitivity. Although clinical studies for DA-9701 have had some limitations, beneficial effects of DA-9701 on FD symptoms and GI functions have been demonstrated in FD patients. Further, the promising results indicate that extended use of DA-9701 for PD patients and functional constipation may be possible. The effective dose of DA-9701, which is well tolerated and safe, seems to be smaller than that of conventional drugs targeting a single pathway. Currently, DA-9701 is a prescription drug for FD in Korea. To be used as a medicine in the global market, further studies involving active compounds are definitely needed for a deeper understanding of the activities of DA-9701 in FD management. For example, efficacy and detailed molecular mechanism studies for each active compound targeting different receptors, alone or in combination for additive, synergistic, or even inhibitory effects, using various FD animal models are needed. Undoubtedly, larger and sophisticatedly designed clinical trials are warranted.

Author Contributions

M.J., M.S. wrote the paper. M.J. had primary responsibility for the final content of the manuscript. All authors read and approved the final manuscript.

Funding

This research was funded by the Bio-Synergy Research Project (NRF-2018M3A9C4076473) of the Ministry of Science and ICT through the National Research Foundation of Korea.

Acknowledgments

We thank Soo Jung Yoon for supporting manuscript preparation.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Brun, R.; Kuo, B. Functional dyspepsia. Ther. Adv. Gastroenterol. 2010, 3, 145–164. [Google Scholar] [CrossRef] [PubMed]
  2. Pasricha, P.J. Neurogastroenterology: A great career choice for aspiring gastroenterologists thinking about the future. Gastroenterology 2011, 140, 1126–1128. [Google Scholar] [CrossRef] [PubMed]
  3. Drossman, D.A. Functional Gastrointestinal Disorders: History, Pathophysiology, Clinical Features, and Rome IV. Gastroenterology 2016, 150, 1262–1279. [Google Scholar] [CrossRef] [PubMed]
  4. Talley, N.J.; Ford, A.C. Functional Dyspepsia. N. Engl. J. Med. 2015, 373, 1853–1863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Lacy, B.E.; Weiser, K.T.; Kennedy, A.T.; Crowell, M.D.; Talley, N.J. Functional dyspepsia: The economic impact to patients. Aliment. Pharmacol. Ther. 2013, 38, 170–177. [Google Scholar] [CrossRef] [PubMed]
  6. Talley, N.J.; Ford, A.C. Functional Dyspepsia. N. Engl. J. Med. 2016, 374, 896. [Google Scholar] [CrossRef] [PubMed]
  7. Koduru, P.; Irani, M.; Quigley, E.M.M. Definition, Pathogenesis, and Management of That Cursed Dyspepsia. Clin. Gastroenterol. Hepatol. 2018, 16, 467–479. [Google Scholar] [CrossRef] [PubMed]
  8. Drossman, D.A. The functional gastrointestinal disorders and the Rome II process. Gut 1999, 45 (Suppl. 2), II1–II5. [Google Scholar] [CrossRef]
  9. Tack, J.; Talley, N.J.; Camilleri, M.; Holtmann, G.; Hu, P.; Malagelada, J.R.; Stanghellini, V. Functional gastroduodenal disorders. Gastroenterology 2006, 130, 1466–1479. [Google Scholar] [CrossRef]
  10. Tack, J.; Talley, N.J. Functional dyspepsia--symptoms, definitions and validity of the Rome III criteria. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 134–141. [Google Scholar] [CrossRef]
  11. Stanghellini, V.; Chan, F.K.; Hasler, W.L.; Malagelada, J.R.; Suzuki, H.; Tack, J.; Talley, N.J. Gastroduodenal Disorders. Gastroenterology 2016, 150, 1380–1392. [Google Scholar] [CrossRef] [PubMed]
  12. Powell, N.; Walker, M.M.; Talley, N.J. Gastrointestinal eosinophils in health, disease and functional disorders. Nat. Rev. Gastroenterol. Hepatol. 2010, 7, 146–156. [Google Scholar] [CrossRef]
  13. Lee, K.J.; Tack, J. Duodenal implications in the pathophysiology of functional dyspepsia. J. Neurogastroenterol. Motil. 2010, 16, 251–257. [Google Scholar] [CrossRef]
  14. Walker, M.M.; Salehian, S.S.; Murray, C.E.; Rajendran, A.; Hoare, J.M.; Negus, R.; Powell, N.; Talley, N.J. Implications of eosinophilia in the normal duodenal biopsy—An association with allergy and functional dyspepsia. Aliment. Pharmacol. Ther. 2010, 31, 1229–1236. [Google Scholar] [CrossRef] [PubMed]
  15. Sugano, K.; Tack, J.; Kuipers, E.J.; Graham, D.Y.; El-Omar, E.M.; Miura, S.; Haruma, K.; Asaka, M.; Uemura, N.; Malfertheiner, P.; et al. Kyoto global consensus report on Helicobacter pylori gastritis. Gut 2015, 64, 1353–1367. [Google Scholar] [CrossRef]
  16. Sarnelli, G.; Caenepeel, P.; Geypens, B.; Janssens, J.; Tack, J. Symptoms associated with impaired gastric emptying of solids and liquids in functional dyspepsia. Am. J. Gastroenterol. 2003, 98, 783–788. [Google Scholar] [CrossRef] [PubMed]
  17. Lee, M.Y. Clinical Relevance of Serotonin Receptor Splice Variant Distribution in Human Colon. J. Neurogastroenterol. Motil. 2015, 21, 303–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. De Maeyer, J.H.; Lefebvre, R.A.; Schuurkes, J.A. 5-HT4 receptor agonists: Similar but not the same. Neurogastroenterol. Motil. 2008, 20, 99–112. [Google Scholar] [CrossRef] [PubMed]
  19. Fayyaz, M.; Lackner, J.M. Serotonin receptor modulators in the treatment of irritable bowel syndrome. Ther. Clin. Risk Manag. 2008, 4, 41–48. [Google Scholar] [PubMed]
  20. Chetty, N.; Coupar, I.M.; Tan, Y.Y.; Desmond, P.V.; Irving, H.R. Distribution of serotonin receptors and interacting proteins in the human sigmoid colon. Neurogastroenterol. Motil. 2009, 21, 551–e15. [Google Scholar] [CrossRef] [PubMed]
  21. Acosta, A.; Camilleri, M. Prokinetics in gastroparesis. Gastroenterol. Clin. N. Am. 2015, 44, 97–111. [Google Scholar] [CrossRef] [PubMed]
  22. Zala, A.V.; Walker, M.M.; Talley, N.J. Emerging drugs for functional dyspepsia. Expert Opin. Emerg. Drugs 2015, 20, 221–233. [Google Scholar] [CrossRef] [PubMed]
  23. Bang, C.S.; Kim, J.H.; Baik, G.H.; Kim, H.S.; Park, S.H.; Kim, E.J.; Kim, J.B.; Suk, K.T.; Yoon, J.H.; Kim, Y.S.; et al. Mosapride treatment for functional dyspepsia: A meta-analysis. J. Gastroenterol. Hepatol. 2015, 30, 28–42. [Google Scholar] [CrossRef] [PubMed]
  24. Pasricha, P.J. Desperately seeking serotonin. A commentary on the withdrawal of tegaserod and the state of drug development for functional and motility disorders. Gastroenterology 2007, 132, 2287–2290. [Google Scholar] [CrossRef] [PubMed]
  25. Huang, X.; Lv, B.; Zhang, S.; Fan, Y.H.; Meng, L.N. Itopride therapy for functional dyspepsia: A meta-analysis. World J. Gastroenterol. 2012, 18, 7371–7377. [Google Scholar] [CrossRef] [PubMed]
  26. Talley, N.J.; Tack, J.; Ptak, T.; Gupta, R.; Giguere, M. Itopride in functional dyspepsia: Results of two phase III multicentre, randomised, double-blind, placebo-controlled trials. Gut 2008, 57, 740–746. [Google Scholar] [CrossRef] [PubMed]
  27. Ehrenpreis, E.D.; Roginsky, G.; Alexoff, A.; Smith, D.G. Domperidone is Commonly Prescribed With QT-Interacting Drugs: Review of a Community-based Practice and a Postmarketing Adverse Drug Event Reporting Database. J. Clin. Gastroenterol. 2017, 51, 56–62. [Google Scholar] [CrossRef]
  28. Ganzini, L.; Casey, D.E.; Hoffman, W.F.; McCall, A.L. The prevalence of metoclopramide-induced tardive dyskinesia and acute extrapyramidal movement disorders. Arch. Intern. Med. 1993, 153, 1469–1475. [Google Scholar] [CrossRef]
  29. Tack, J.; Janssen, P.; Masaoka, T.; Farre, R.; Van Oudenhove, L. Efficacy of buspirone, a fundus-relaxing drug, in patients with functional dyspepsia. Clin. Gastroenterol. Hepatol. 2012, 10, 1239–1245. [Google Scholar] [CrossRef]
  30. Sekino, Y.; Yamada, E.; Sakai, E.; Ohkubo, H.; Higurashi, T.; Iida, H.; Endo, H.; Takahashi, H.; Koide, T.; Sakamoto, Y.; et al. Influence of sumatriptan on gastric accommodation and on antral contraction in healthy subjects assessed by ultrasonography. Neurogastroenterol. Motil. 2012, 24, 1083–e564. [Google Scholar] [CrossRef]
  31. Tack, J.; Janssen, P. Acotiamide (Z-338, YM443), a new drug for the treatment of functional dyspepsia. Expert Opin. Investig. Drugs 2011, 20, 701–712. [Google Scholar] [CrossRef]
  32. Kusunoki, H.; Haruma, K.; Manabe, N.; Imamura, H.; Kamada, T.; Shiotani, A.; Hata, J.; Sugioka, H.; Saito, Y.; Kato, H.; et al. Therapeutic efficacy of acotiamide in patients with functional dyspepsia based on enhanced postprandial gastric accommodation and emptying: Randomized controlled study evaluation by real-time ultrasonography. Neurogastroenterol. Motil. 2012, 24, 540–e251. [Google Scholar] [CrossRef]
  33. Matsueda, K.; Hongo, M.; Tack, J.; Saito, Y.; Kato, H. A placebo-controlled trial of acotiamide for meal-related symptoms of functional dyspepsia. Gut 2012, 61, 821–828. [Google Scholar] [CrossRef] [PubMed]
  34. Coffin, B.; Azpiroz, F.; Guarner, F.; Malagelada, J.R. Selective gastric hypersensitivity and reflex hyporeactivity in functional dyspepsia. Gastroenterology 1994, 107, 1345–1351. [Google Scholar] [CrossRef]
  35. Bradette, M.; Pare, P.; Douville, P.; Morin, A. Visceral perception in health and functional dyspepsia. Crossover study of gastric distension with placebo and domperidone. Dig. Dis. Sci. 1991, 36, 52–58. [Google Scholar] [CrossRef] [PubMed]
  36. Mearin, F.; Cucala, M.; Azpiroz, F.; Malagelada, J.R. The origin of symptoms on the brain-gut axis in functional dyspepsia. Gastroenterology 1991, 101, 999–1006. [Google Scholar] [CrossRef]
  37. Tack, J.; Caenepeel, P.; Fischler, B.; Piessevaux, H.; Janssens, J. Symptoms associated with hypersensitivity to gastric distention in functional dyspepsia. Gastroenterology 2001, 121, 526–535. [Google Scholar] [CrossRef] [PubMed]
  38. Farre, R.; Vanheel, H.; Vanuytsel, T.; Masaoka, T.; Tornblom, H.; Simren, M.; Van Oudenhove, L.; Tack, J.F. In functional dyspepsia, hypersensitivity to postprandial distention correlates with meal-related symptom severity. Gastroenterology 2013, 145, 566–573. [Google Scholar] [CrossRef] [PubMed]
  39. Thompson, A.J.; Lummis, S.C. The 5-HT3 receptor as a therapeutic target. Expert Opin. Ther. Targets 2007, 11, 527–540. [Google Scholar] [CrossRef] [PubMed]
  40. Delvaux, M.; Louvel, D.; Mamet, J.P.; Campos-Oriola, R.; Frexinos, J. Effect of alosetron on responses to colonic distension in patients with irritable bowel syndrome. Aliment. Pharmacol. Ther. 1998, 12, 849–855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Harris, L.A.; Chang, L. Alosetron: An effective treatment for diarrhea-predominant irritable bowel syndrome. Womens Health 2007, 3, 15–27. [Google Scholar] [CrossRef] [PubMed]
  42. Pascaud, X.; Honde, C.; Le Gallou, B.; Chanoine, F.; Roman, F.; Bueno, L.; Junien, J.L. Effects of fedotozine on gastrointestinal motility in dogs: Mechanism of action and related pharmacokinetics. J. Pharm. Pharmacol. 1990, 42, 546–552. [Google Scholar] [CrossRef] [PubMed]
  43. Diop, L.; Riviere, P.J.; Pascaud, X.; Junien, J.L. Peripheral kappa-opioid receptors mediate the antinociceptive effect of fedotozine (correction of fetodozine) on the duodenal pain reflex inrat. Eur J. Pharmacol. 1994, 271, 65–71. [Google Scholar] [CrossRef]
  44. De Ponti, F. Drug development for the irritable bowel syndrome: Current challenges and future perspectives. Front. Pharmacol. 2013, 4, 7. [Google Scholar] [CrossRef] [PubMed]
  45. Talley, N.J.; American Gastroenterological, A. American Gastroenterological Association medical position statement: Evaluation of dyspepsia. Gastroenterology 2005, 129, 1753–1755. [Google Scholar] [CrossRef] [PubMed]
  46. Talley, N.J.; Locke, G.R.; Saito, Y.A.; Almazar, A.E.; Bouras, E.P.; Howden, C.W.; Lacy, B.E.; DiBaise, J.K.; Prather, C.M.; Abraham, B.P.; et al. Effect of Amitriptyline and Escitalopram on Functional Dyspepsia: A Multicenter, Randomized Controlled Study. Gastroenterology 2015, 149, 340–349. [Google Scholar] [CrossRef]
  47. Vanheel, H.; Tack, J. Therapeutic options for functional dyspepsia. Dig. Dis. 2014, 32, 230–234. [Google Scholar] [CrossRef]
  48. Tack, J.; Ly, H.G.; Carbone, F.; Vanheel, H.; Vanuytsel, T.; Holvoet, L.; Boeckxstaens, G.; Caenepeel, P.; Arts, J.; Van Oudenhove, L. Efficacy of Mirtazapine in Patients With Functional Dyspepsia and Weight Loss. Clin. Gastroenterol. Hepatol. 2016, 14, 385–392. [Google Scholar] [CrossRef]
  49. Choung, R.S.; Cremonini, F.; Thapa, P.; Zinsmeister, A.R.; Talley, N.J. The effect of short-term, low-dose tricyclic and tetracyclic antidepressant treatment on satiation, postnutrient load gastrointestinal symptoms and gastric emptying: A double-blind, randomized, placebo-controlled trial. Neurogastroenterol. Motil. 2008, 20, 220–227. [Google Scholar] [CrossRef]
  50. Mansi, C.; Borro, P.; Giacomini, M.; Biagini, R.; Mele, M.R.; Pandolfo, N.; Savarino, V. Comparative effects of levosulpiride and cisapride on gastric emptying and symptoms in patients with functional dyspepsia and gastroparesis. Aliment. Pharmacol. Ther. 2000, 14, 561–569. [Google Scholar] [CrossRef]
  51. Mearin, F.; Rodrigo, L.; Perez-Mota, A.; Balboa, A.; Jimenez, I.; Sebastian, J.J.; Paton, C. Levosulpiride and cisapride in the treatment of dysmotility-like functional dyspepsia: A randomized, double-masked trial. Clin. Gastroenterol. Hepatol. 2004, 2, 301–308. [Google Scholar] [CrossRef]
  52. Camilleri, M.; Bueno, L.; de Ponti, F.; Fioramonti, J.; Lydiard, R.B.; Tack, J. Pharmacological and pharmacokinetic aspects of functional gastrointestinal disorders. Gastroenterology 2006, 130, 1421–1434. [Google Scholar] [CrossRef] [PubMed]
  53. Kwon, Y.S.; Son, M. DA-9701: A New Multi-Acting Drug for the Treatment of Functional Dyspepsia. Biomol. Ther. 2013, 21, 181–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Ding, B.; Zhou, T.; Fan, G.; Hong, Z.; Wu, Y. Qualitative and quantitative determination of ten alkaloids in traditional Chinese medicine Corydalis yanhusuo W.T. Wang by LC-MS/MS and LC-DAD. J. Pharm. Biomed. Anal. 2007, 45, 219–226. [Google Scholar] [CrossRef]
  55. Kumar, S.; Kumar, D.; Singh, J.; Narender, R.; Kaushik, D. Anti-inflammatory, Analgesic and Antioxidant Activities of Ipomoea hederaceae Linn. Planta Med. 2009, 75, p-100. [Google Scholar] [CrossRef]
  56. Soji, Y.; Kadokawa, T.; Masuda, Y.; Kawashima, K.; Nakamura, K. Effects of Corydalis alkaloid upon inhibition of gastric juice secretion and prevention of gastric ulcer in experimental animals. Nihon Yakurigaku Zasshi 1969, 65, 196–209. [Google Scholar] [CrossRef]
  57. Kamigauchi, M.; Iwasa, K. Corydalis spp.: In Vitro Culture and the Biotransformation of Protoberberines. Biotechnol. Agric. For. 1994, 26, 93–105. [Google Scholar] [CrossRef]
  58. Yun, K.J.; Shin, J.S.; Choi, J.H.; Back, N.I.; Chung, H.G.; Lee, K.T. Quaternary alkaloid, pseudocoptisine isolated from tubers of Corydalis turtschaninovi inhibits LPS-induced nitric oxide, PGE2, and pro-inflammatory cytokines production via the down-regulation of NF-kappaB in RAW 264.7 murine macrophage cells. Int. Immunopharmacol. 2009, 9, 1323–1331. [Google Scholar] [CrossRef]
  59. Jung, J.W.; Kim, J.M.; Jeong, J.S.; Son, M.; Lee, H.S.; Lee, M.G.; Kang, H.E. Pharmacokinetics of chlorogenic acid and corydaline in DA-9701, a new botanical gastroprokinetic agent, in rats. Xenobiotica 2014, 44, 635–643. [Google Scholar] [CrossRef]
  60. Lee, T.H.; Choi, J.J.; Kim, D.H.; Choi, S.; Lee, K.R.; Son, M.; Jin, M. Gastroprokinetic effects of DA-9701, a new prokinetic agent formulated with Pharbitis Semen and Corydalis Tuber. Phytomedicine 2008, 15, 836–843. [Google Scholar] [CrossRef]
  61. Choi, S.; Choi, J.J.; Jun, J.Y.; Koh, J.W.; Kim, S.H.; Kim, D.H.; Pyo, M.Y.; Choi, S.; Son, J.P.; Lee, I.; et al. Induction of pacemaker currents by DA-9701, a prokinetic agent, in interstitial cells of Cajal from murine small intestine. Mol. Cells 2009, 27, 307–312. [Google Scholar] [CrossRef] [PubMed]
  62. Lim, C.H.; Choi, M.G.; Park, H.; Baeg, M.K.; Park, J.M. Effect of DA-9701 on gastric emptying in a mouse model: Assessment by 13C-octanoic acid breath test. World J. Gastroenterol. 2013, 19, 4380–4385. [Google Scholar] [CrossRef] [PubMed]
  63. Kang, J.W.; Han, D.K.; Kim, O.N.; Lee, K.J. Effect of DA-9701 on the Normal Motility and Clonidine-induced Hypomotility of the Gastric Antrum in Rats. J. Neurogastroenterol. Motil. 2016, 22, 304–309. [Google Scholar] [CrossRef] [Green Version]
  64. Hussain, Z.; Rhee, K.W.; Lee, Y.J.; Park, H. The Effect of DA-9701 in Opioid-induced Bowel Dysfunction of Guinea Pig. J. Neurogastroenterol. Motil. 2016, 22, 529–538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Lee, S.P.; Lee, O.Y.; Lee, K.N.; Lee, H.L.; Choi, H.S.; Yoon, B.C.; Jun, D.W. Effect of DA-9701, a Novel Prokinetic Agent, on Post-operative Ileus in Rats. J. Neurogastroenterol. Motil. 2017, 23, 109–116. [Google Scholar] [CrossRef] [Green Version]
  66. Jo, S.Y.; Hussain, Z.; Lee, Y.J.; Park, H. Corticotrophin-releasing factor-mediated effects of DA-9701 in Postoperative Ileus Guinea Pig Model. Neurogastroenterol. Motil. 2018, 30, e13385. [Google Scholar] [CrossRef]
  67. Jung, Y.S.; Kim, M.Y.; Lee, H.S.; Park, S.L.; Lee, K.J. Effect of DA-9701, a novel prokinetic agent, on stress-induced delayed gastric emptying and hormonal changes in rats. Neurogastroenterol. Motil. 2013, 25, 254–e166. [Google Scholar] [CrossRef]
  68. Lee, T.H.; Son, M.; Kim, S.Y. Effects of corydaline from Corydalis tuber on gastric motor function in an animal model. Biol. Pharm. Bull. 2010, 33, 958–962. [Google Scholar] [CrossRef]
  69. Kim, E.R.; Min, B.H.; Lee, S.O.; Lee, T.H.; Son, M.; Rhee, P.L. Effects of DA-9701, a novel prokinetic agent, on gastric accommodation in conscious dogs. J. Gastroenterol. Hepatol. 2012, 27, 766–772. [Google Scholar] [CrossRef]
  70. Kim, Y.S.; Lee, M.Y.; Park, J.S.; Choi, E.S.; Kim, M.S.; Park, S.H.; Ryu, H.S.; Choi, S.C. Effect of DA-9701 on Feeding Inhibition Induced by Acute Restraint Stress in Rats. Korean J. Helicobacter Up. Gastrointest. Res. 2018, 18, 50–55. [Google Scholar] [CrossRef]
  71. Lee, T.H.; Kim, K.H.; Lee, S.O.; Lee, K.R.; Son, M.; Jin, M. Tetrahydroberberine, an isoquinoline alkaloid isolated from corydalis tuber, enhances gastrointestinal motor function. J. Pharmacol. Exp. Ther. 2011, 338, 917–924. [Google Scholar] [CrossRef] [PubMed]
  72. Oh, K.H.; Nam, Y.; Jeong, J.H.; Kim, I.K.; Sohn, U.D. The effect of DA-9701 on 5-hydroxytryptamine-induced contraction of feline esophageal smooth muscle cells. Molecules 2014, 19, 5135–5149. [Google Scholar] [CrossRef] [PubMed]
  73. Min, Y.W.; Ko, E.J.; Lee, J.Y.; Min, B.H.; Lee, J.H.; Kim, J.J.; Rhee, P.L. Nitrergic Pathway Is the Major Mechanism for the Effect of DA-9701 on the Rat Gastric Fundus Relaxation. J. Neurogastroenterol. Motil. 2014, 20, 318–325. [Google Scholar] [CrossRef] [Green Version]
  74. Kim, E.R.; Min, B.H.; Lee, T.H.; Son, M.; Rhee, P.L. Effect of DA-9701 on colorectal distension-induced visceral hypersensitivity in a rat model. Gut Liver 2014, 8, 388–393. [Google Scholar] [CrossRef] [PubMed]
  75. Cao, F.L.; Shang, G.W.; Wang, Y.; Yang, F.; Li, C.L.; Chen, J. Antinociceptive effects of intragastric DL-tetrahydropalmatine on visceral and somatic persistent nociception and pain hypersensitivity in rats. Pharmacol. Biochem. Behav. 2011, 100, 199–204. [Google Scholar] [CrossRef] [PubMed]
  76. Lee, S.P.; Lee, K.N.; Lee, O.Y.; Lee, H.L.; Jun, D.W.; Yoon, B.C.; Choi, H.S.; Hwang, S.J.; Lee, S.E. Effects of DA-9701, a novel prokinetic agent, on phosphorylated extracellular signal-regulated kinase expression in the dorsal root ganglion and spinal cord induced by colorectal distension in rats. Gut Liver 2014, 8, 140–147. [Google Scholar] [CrossRef] [PubMed]
  77. Ramsbottom, N.; Hunt, J.N. Studies of the effect of metoclopramide and apomorphine on gastric emptying and secretion in man. Gut 1970, 11, 989–993. [Google Scholar] [CrossRef] [Green Version]
  78. Tanila, H.; Kauppila, T.; Taira, T. Inhibition of intestinal motility and reversal of postlaparotomy ileus by selective alpha 2-adrenergic drugs in the rat. Gastroenterology 1993, 104, 819–824. [Google Scholar] [CrossRef]
  79. Hirokawa, Y.; Yamazaki, H.; Yoshida, N.; Kato, S. A novel series of 6-methoxy-1H-benzotriazole-5-carboxamide derivatives with dual antiemetic and gastroprokinetic activities. Bioorg. Med. Chem. Lett. 1998, 8, 1973–1978. [Google Scholar] [CrossRef]
  80. Haug, T.T.; Svebak, S.; Wilhelmsen, I.; Berstad, A.; Ursin, H. Psychological factors and somatic symptoms in functional dyspepsia. A comparison with duodenal ulcer and healthy controls. J. Psychosom. Res. 1994, 38, 281–291. [Google Scholar] [CrossRef]
  81. Nakade, Y.; Tsuchida, D.; Fukuda, H.; Iwa, M.; Pappas, T.N.; Takahashi, T. Restraint stress delays solid gastric emptying via a central CRF and peripheral sympathetic neuron in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2005, 288, R427–R432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Tache, Y.; Martinez, V.; Million, M.; Wang, L. Stress and the gastrointestinal tract III. Stress-related alterations of gut motor function: Role of brain corticotropin-releasing factor receptors. Am. J. Physiol. Gastrointest. Liver Physiol. 2001, 280, G173–G177. [Google Scholar] [CrossRef] [PubMed]
  83. Lim, H.C.; Park, H.; Lee, S.I.; Jahng, J.; Lee, Y.J. Tu1985 effects of DA-9701, a novel prokinetic agent on gastric motor function in guinea pig. Gastroenterology 2013, 142, S-893. [Google Scholar] [CrossRef]
  84. Hansen, M.B. Neurohumoral control of gastrointestinal motility. Physiol. Res. 2003, 52, 1–30. [Google Scholar] [PubMed]
  85. Azpiroz, F.; Malagelada, J.R. Abdominal bloating. Gastroenterology 2005, 129, 1060–1078. [Google Scholar] [CrossRef]
  86. Azpiroz, F.; Malagelada, J.R. Physiological variations in canine gastric tone measured by an electronic barostat. Am. J. Physiol. 1985, 248, G229–G237. [Google Scholar] [CrossRef] [PubMed]
  87. De Ponti, F.; Crema, F.; Moro, E.; Nardelli, G.; Frigo, G.; Crema, A. Role of 5-HT1B/D receptors in canine gastric accommodation: Effect of sumatriptan and 5-HT1B/D receptor antagonists. Am. J. Physiol. Gastrointest. Liver Physiol. 2003, 285, G96–G104. [Google Scholar] [CrossRef] [PubMed]
  88. Azpiroz, F.; Malagelada, J.R. Vagally mediated gastric relaxation induced by intestinal nutrients in the dog. Am. J. Physiol. 1986, 251, G727–G735. [Google Scholar] [CrossRef] [PubMed]
  89. Kuiken, S.D.; Tytgat, G.N.; Boeckxstaens, G.E. Review article: Drugs interfering with visceral sensitivity for the treatment of functional gastrointestinal disorders--the clinical evidence. Aliment. Pharmacol. Ther. 2005, 21, 633–651. [Google Scholar] [CrossRef]
  90. Coulie, B.; Tack, J.; Sifrim, D.; Andrioli, A.; Janssens, J. Role of nitric oxide in fasting gastric fundus tone and in 5-HT1 receptor-mediated relaxation of gastric fundus. Am. J. Physiol. 1999, 276, G373–G377. [Google Scholar] [CrossRef]
  91. Bonnet, F.; Boico, O.; Rostaing, S.; Loriferne, J.F.; Saada, M. Clonidine-induced analgesia in postoperative patients: Epidural versus intramuscular administration. Anesthesiology 1990, 72, 423–427. [Google Scholar] [CrossRef] [PubMed]
  92. Mayer, E.A.; Gebhart, G.F. Basic and clinical aspects of visceral hyperalgesia. Gastroenterology 1994, 107, 271–293. [Google Scholar] [CrossRef]
  93. Camilleri, M.; Coulie, B.; Tack, J.F. Visceral hypersensitivity: Facts, speculations, and challenges. Gut 2001, 48, 125–131. [Google Scholar] [CrossRef] [PubMed]
  94. Choi, M.G.; Rhee, P.L.; Park, H.; Lee, O.Y.; Lee, K.J.; Choi, S.C.; Seol, S.Y.; Chun, H.J.; Rew, J.S.; Lee, D.H.; et al. Randomized, Controlled, Multi-center Trial: Comparing the Safety and Efficacy of DA-9701 and Itopride Hydrochloride in Patients With Functional Dyspepsia. J. Neurogastroenterol. Motil. 2015, 21, 414–422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Jung, H.K.; Lee, K.J.; Choi, M.G.; Park, H.; Lee, J.S.; Rhee, P.L.; Kim, N.; Park, K.; Choi, S.C.; Lee, O.Y.; et al. Efficacy of DA-9701 (Motilitone) in Functional Dyspepsia Compared to Pantoprazole: A Multicenter, Randomized, Double-blind, Non-inferiority Study. J. Neurogastroenterol. Motil. 2016, 22, 254–263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Park, C.H.; Kim, H.S.; Lee, S.K. Effects of the New Prokinetic Agent DA-9701 Formulated With Corydalis Tuber and Pharbitis Seed in Patients With Minimal Change Esophagitis: A Bicenter, Randomized, Double Blind, Placebo-controlled Study. J. Neurogastroenterol. Motil. 2014, 20, 338–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Shin, C.M.; Lee, Y.J.; Kim, J.M.; Lee, J.Y.; Kim, K.J.; Choi, Y.J.; Kim, N.; Lee, D.H. DA-9701 on gastric motility in patients with Parkinson’s disease: A randomized controlled trial. Parkinsonism Relat. Disord. 2018, 54, 84–89. [Google Scholar] [CrossRef] [PubMed]
  98. Min, Y.W.; Min, B.H.; Kim, S.; Choi, D.; Rhee, P.L. Effect of DA-9701 on Gastric Motor Function Assessed by Magnetic Resonance Imaging in Healthy Volunteers: A Randomized, Double-Blind, Placebo-Controlled Trial. PLoS ONE 2015, 10, e0138927. [Google Scholar] [CrossRef]
  99. Kim, S.Y.; Woo, H.S.; Kim, K.O.; Choi, S.H.; Kwon, K.A.; Chung, J.W.; Kim, Y.J.; Kim, J.H.; Kim, S.J.; Park, D.K. DA-9701 improves colonic transit time and symptoms in patients with functional constipation: A prospective study. J. Gastroenterol. Hepatol. 2017, 32, 1943–1948. [Google Scholar] [CrossRef]
Figure 1. Major drug targets associated with pathophysiology of functional dyspepsia and current drugs. Targets of DA-9701 are presented.
Figure 1. Major drug targets associated with pathophysiology of functional dyspepsia and current drugs. Targets of DA-9701 are presented.
Ijms 19 04035 g001
Table 1. Pharmacological profile summary of DA 9701.
Table 1. Pharmacological profile summary of DA 9701.
ClassStudy TypeExperimentalResultsRef.
Prokinetics
D2 antagonistic activityIn vivoRat, gastric emptying delayed by apomorphine and cisplatin treatmentAcceleration of gastric emptying in normal rats, improvement in delayed gastric emptying[60]
Gastroprokinetic activityIn vitroWhole cell patch clampModulation of pacemaker activity[61]
Gastroprokinetic activityIn vivoMouse, 13C-octanoic acid breath testAcceleration of gastric emptying[62]
Gastroprokinetic activityIn vivoRat, clonidine-induced hypomotility of the gastric antrumDA-9701 improved the clonidine-induced hypomotility of the gastric antrum[63]
Gastroprokinetic activityIn vivoMouse, laparotomy or atropine injectionEnhancement of gastrointestinal transit[60]
Gastroprokinetic activityIn vivoGuinea pig, opioid-induced bowel dysfunctionIncrease in amplitude of ileal muscle contraction, restoration of delayed GI transit[64]
Gastroprokinetic activityIn vivoRat, postoperative ileus (POI)Amelioration of POI by reduction in delayed GIT, improvement in defecation in rat model of POI[65]
Prokinetic activityIn vivoGuinea pig, postoperative ileus (POI)Improvement in GI transit, inhibition of plasma ACTH levels via central CRF pathways[66]
Gastroprokinetic activityIn vivoRat, stress-induced delayed gastric emptyingImprovement in delayed gastric emptying and inhibition of the hormonal changes induced by stress[67]
Fundic relaxants
Fundus-relaxing activityIn vivoBeagle dog, canine gastric compliance with barostatInduction of gastric relaxation and increased gastric compliance[60]
Fundus-relaxing activityIn vivoBeagle dog, canine gastric compliance with barostatCD from Corydalis Tuber induced gastric relaxation and facilitated gastric accommodation[68]
Fundus-relaxing activityIn vivoBeagle dog, meal-induced gastric accommodation with barostatImprovement in gastric accommodation via increase in postprandial gastric volume[69]
5-HT1A agonist activityIn vivoRat, restraint stress-induced feeding inhibitionDA-9701 was blocked by the 5-HT1A antagonist, WAY 100635[70]
D2 antagonistic activity, 5-HT1A agonist activityIn vivoRat, restraint stress-induced impaired gastric complianceTetrahydroberberine, an isoquinoline alkaloid isolated from Corydalis Tuber, enhanced gastric accommodation[71]
D2 antagonistic activity, 5-HT1A agonist activityIn vivoBeagle dog, canine gastric compliance with barostatTetrahydroberberine, an isoquinoline alkaloid isolated from Corydalis Tuber, enhanced gastric accommodation[71]
Down-regulation of 5-HT-induced contractionIn vitroFeline, esophageal smooth muscle cellsInhibition of 5-HT-induced contraction via inhibition of MLC20 phosphorylation[72]
Gastric fundus relaxationIn vivoRat, electrical field stimulation (EFS)-induced contractile responsesNitrergic pathway is major mechanism involved in relaxation of rat gastric fundus by DA-9701[73]
Visceral hypersensitivity
Antinociceptive effectIn vivoNeonatal rat colorectal distension (CRD)-induced visceral hypersensitivitySignificant decrease in mean arterial pressure (MAP) changes after CRD[74]
Visceral pain modulationIn vivoRat, bee venom (BV)-induced persistent spontaneous nociception and pain hypersensitivityTetrahydropalmatine, an alkaloid constituent of plants from the genera Stephania and Corydalis effectively inhibited visceral nociception as well as thermal and mechanical inflammatory pain hypersensitivity[75]
Visceral pain modulationIn vivoRat, CRD-induced visceral painDecrease in visceral pain via reduction in p-ERK in the dorsal root ganglion and spinal cord[76]
Table 2. Clinical trials using DA-9701.
Table 2. Clinical trials using DA-9701.
DiseaseTreatmentStudy DesignSample SizeEnd PointSign(Ref.)
FD, Rome IIDA 9701 vs. itoprideRandomized, double-blind464 patientsChange from baseline in the composite score of the 8 dyspeptic symptoms. Overall treatment effect (OTE) as rated on a 7-grade scale.Significant improvement in symptoms and non-inferior efficacy and comparable safety to that of itopride[94]
FD, Rome IIIDA 9701 vs. pantoprazoleRandomized, double-blind389 patientsGlobal symptom assessment, response rates, difference in each score and total score of FD symptoms, difference in dyspepsia-specific quality of life (QOL) outcomes, symptomatic relief according to the subtypes of FD, i.e., epigastric pain syndrome (EPS) and postprandial distress syndrome (PDS).Improvement in global and individual symptoms and increase in dyspepsia-specific QOL among patients. Efficacy of the monotherapy was comparable to that of pantoprazole. There was no additive effect of the combination of DA-9701 and pantoprazole.[95]
Minimal change esophagitisDA 9701 vs. PlaceboDouble blind, placebo-controlled 81 patientsChanges in Nepean dyspepsia index questionnaire-Korean version (NDI-K) symptom scoreAlthough NDI-K symptom scores and QOL scores improved after 4 weeks of treatment compared to baseline values in patients with minimal change esophagitis, DA-9701 did not improve the symptom scores or QOL scores compared to those of placebo.[96]
Parkinson’s disease (PD)DA 9701 vs. domperidoneRandomized, double-blind40 patientsGastric MRI, laboratory testingUsed for patients with PD to enhance gastric motility without aggravating PD symptoms [97]
Healthy volunteersDA 9701 vs. placeboRandomized, double-blind, placebo-controlled40 healthy volunteersGastric MRIDA-9701 enhanced gastric emptying and did not significantly affect gastric accommodation in healthy volunteers.[98]
Functional constipation, Rome IIIDA 9701Prospective study37 patientsColonic transit time (CTT) measurementDA-9701 accelerated colonic transit and safely improved symptoms [99]

Share and Cite

MDPI and ACS Style

Jin, M.; Son, M. DA-9701 (Motilitone): A Multi-Targeting Botanical Drug for the Treatment of Functional Dyspepsia. Int. J. Mol. Sci. 2018, 19, 4035. https://doi.org/10.3390/ijms19124035

AMA Style

Jin M, Son M. DA-9701 (Motilitone): A Multi-Targeting Botanical Drug for the Treatment of Functional Dyspepsia. International Journal of Molecular Sciences. 2018; 19(12):4035. https://doi.org/10.3390/ijms19124035

Chicago/Turabian Style

Jin, Mirim, and Miwon Son. 2018. "DA-9701 (Motilitone): A Multi-Targeting Botanical Drug for the Treatment of Functional Dyspepsia" International Journal of Molecular Sciences 19, no. 12: 4035. https://doi.org/10.3390/ijms19124035

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop