Minireviews Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Oct 7, 2015; 21(37): 10644-10653
Published online Oct 7, 2015. doi: 10.3748/wjg.v21.i37.10644
Are probiotics useful in Helicobacter pylori eradication?
Matjaž Homan, Rok Orel, University Children’s Hospital, 1000 Ljubljana, Slovenia
Author contributions: Homan M was responsible for literature review and wrote the paper; Orel R was responsible for the critically revising the paper and making important suggestions.
Conflict-of-interest statement: The authors have no conflict of interest to declare.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Matjaž Homan, MD, PhD, University Children’s Hospital, Bohoričeva 20, 1000 Ljubljana, Slovenia. matjaz.homan@guest.arnes.si
Telephone: + 386-15229276 Fax: + 386-15229206
Received: February 28, 2015
Peer-review started: March 3, 2015
First decision: March 30, 2015
Revised: April 25, 2015
Accepted: September 2, 2015
Article in press: September 2, 2015
Published online: October 7, 2015

Abstract

Helicobacter pylori (H. pylori) is considered an etiologic factor for the development of peptic ulcer disease, gastric adenocarcinoma, and MALT lymphoma. Therapeutic schemes to eradicate the bacteria are based on double antibiotic therapy and proton pump inhibitor. Despite many therapeutic improvements in H. pylori eradication treatment, it is still associated with high infection rate also in developed countries. Bacterial resistance and adverse events occurrence are among most frequent causes for anti- H. pylori treatment failure. Several studies have reported that certain probiotic strains can exhibit inhibitory activity against H. pylori bacteria. In addition, some probiotic strains can reduce the occurrence of side effects due to antibiotic therapy and consequently increase the H. pylori eradication rate. The results of the prospective double-blind placebo-controlled studies suggest that specific probiotics, such as S. boulardii and L. johnsonni La1 probably can diminish the bacterial load, but not completely eradicate the H. pylori bacteria. Furthermore, it seems that supplementation with S. boulardii is a useful concomitant therapy in the standard H. pylori eradication treatment protocol and most probably increases eradication rate. L. reuteri is equally effective, but more positive studies are needed. Finally, probiotic strains, such as S. boulardii, L. reuteri and L. GG, decrease gastrointestinal antibiotic associated adverse effects.

Key Words: Helicobacter pylori, Probiotics, Eradication therapy, Adverse effects, Strain

Core tip: Poor eradication rates in Helicobacter pylori (H. pylori) infected patients have been reported, which was mostly explained by the increased rates of bacterial resistance to antibiotics and a low compliance for those drugs. This situation needs the development of alternative treatment options for the H. pylori infection in patients. The results of recent studies suggest that certain probiotic strains supplemented to standard eradication therapy diminish the frequency of gastrointestinal adverse effects and consequently also increase the eradication rates.



HELICOBACTER PYLORI

Helicobacter pylori (H. pylori) is a Gram-negative micro-organism. From the original discovery of genus Helicobacter, more than 20 species have now been formally defined[1]. The gastric mucosa is well protected against bacterial infection. After ingestion H. pylori must survive the bactericidal activity of the gastric lumen and enter the mucus layer. H. pylori is highly adapted to special conditions in the stomach. It possess flagella, which enable colonization of the gastric epithelium[2]. Moreover, the bacteria produce enzyme urease, which hydrolyzes urea into carbon dioxide and ammonia and elevates pH in the surroundings of the bacteria. The enzyme activity is highest at low pH[3]. The H. pylori bacteria usually causes chronic infection due to a complex balance between host factors and virulence bacterial factors. Among several bacterial factors one of the main factors, which drive Th17 inflammation, represents the secreted peptidyl prolyl cis, trans-isomerase[4]. In the colonization and chronic infection the adherence of the bacteria to the gastric epithelium is also very important. Lipopolysaccharide is a component of bacterial wall consisted of an O-specific polysaccharide chain, oligosaccharide, and the lipid A. The Lewis-like antigens (Lex , Ley, Lea, Leb) of H. pylori lipopolysaccharide are also expressed in the epithelial cells of gastric mucosa, therefore they probably play an important role in adherence to gastric cells[5]. Around 5% of the H. pylori genome encodes outer membrane proteins (OMP). OMP are adhesins on the surface of the bacteria, which promote binding to the epithelial cells in the stomach. The major OMP is the blood group antigen-binding adhesion A (BabA), which mediates the binding of H. pylori to the fucosylated Leb blood group antigen. It also mediates binding to salivary mucin MUC5B, a proline-rich glycoprotein, and to the glycoprotein gp-340[6]. The second most important OMP is the sialic acid-binding adhesion (SabA)[7]. SabA mediates the binding of the bacteria to the sialyl-dimeric-Lex, to salivary mucin MUC5B, and to salivary glycoproteins like MUC7 and zinc-glycoprotein[8]. In gastric colonisation are also involved other OMPs like AlpA, AlpB, HopZ and HomB. Tight binding of bacteria to gastric epithelial cells is enabled therefore also with BabA2 and Saba adhesins. The H. pylori is very closely associated with extracellular MUC5AC and epithelial cells that produce MUC5AC, therefore MUC5AC plays a role in the adherence of H. pylori to the gastric mucosa. The important receptor for bacteria is the Leb structure present in the normal gastric tissue and MUC5AC is the most important carrier of Leb, with the attachment being made through BabA[9].

H. pylori infection is still one of the most common bacterial infections all over the world. H. pylori infection is very common in Eastern Europe, Africa and most Asian countries[10]. In developed parts of the world the prevalence has lowered and is below 10% in children and below 30% in adults[11].

Twenty percent of infected individuals develop symptomatic gastritis, gastric or duodenal ulcer, gastric adenocarcinoma, and non-Hodgkin’s gastric lymphoma[12]. H. pylori is also associated with iron-deficiency anemia, idiopathic thrombocytopenic purpura and vitamin B12 deficiency[13]. Several bacterial, host and environmental factors have been studied to determine clinical outcome of H. pylori infection. Among bacterial factors, virulence genes are most important, and the severity of H. pylori related disease correlates with the presence of cagA, vacA s1m1 and babA2 genotypes[14,15]. Chronic active gastritis can proceed to precancerous lesions such as gastric mucosal atrophy and intestinal metaplasia, and finally to the development of gastric adenocarcinoma. The results of recent studies support the beneficial effect of H. pylori eradication on preventing gastric cancer as well as on the regression of mucosal atrophy and intestinal metaplasia of the gastric mucosa[16]. Therefore, precancerous gastric lesions demand rapid detection of H. pylori and eradication of bacteria.

H. pylori is suggested to have also beneficial properties. Several studies and meta- analysis showed an inverse relationship between H. pylori infection and asthma occurrence[17,18]. The association was stronger for children than adults, but more prospective studies are needed to confirm the above mention relationship. In addition, H. pylori infection probably decreases the prevalence of obesity in children[19]. The association between H. pylori infection and gastroesophageal reflux disease is still unclear. Although it has previously been suggested that H. pylori eradication may cause reflux disease, the existence of such association was not confirmed recently[20].

The standard eradication therapy consists of two antibiotics and a proton pump inhibitor lasting for 7-14 d. The percentage of treatment failures is rising and major cause for this is bacterial resistance to frequently prescribed antibiotics. The frequent use of clarithromycin for respiratory tract infections has led to high H. pylori clarithromycin resistance rates[21]. Resistance to metronidazole has less clinical impact. Metronidazole resistance can be partially overcome by increasing the dose and treatment duration. The resistance of H. pylori to metronidazole has been reported between 30% and 40%[22]. The eradication levels using standard triple therapy is between 60% and 80%, the last being regarded as the minimal acceptable level according to the Maastricht guidelines[23]. Therefore, there is a great interest of developing new alternatives to eradication therapy, such as quadruple therapy or sequential therapy. Another possibility is to add the additional drug to standard therapy protocol. Several studies have examined the potential influence of probiotics as adjuncts to standard therapy on H. pylori eradication rate.

In addition, probiotics were studied to lower the frequency of side effects, because adverse events relating to H. pylori therapy are an important factor that influences compliance. The overall rate of adverse events was 53.3% in a multicentre study[24]. The most common adverse events reported are diarrhoea, nausea and vomiting, which have significant physical and social impacts, and it has been shown that side effects were significantly associated with decreased compliance and treatment failure[25].

PROBIOTICS

The FAO/WHO definition of probiotics is that probiotics are live microorganisms which when administered in adequate amounts confer a health benefit on the host[26]. The most commonly used probiotics in humans are microorganisms belonging to genera Bififobacterium, Lactobacillus, Saccharomyces and Bacillus[27]. They are used as single species or multispecies preparation. The beneficial effects of probiotics seem to be strain-specific and dose-dependent. Clinical studies of probiotics in human are rapidly increasing. The beneficial effect of probiotics is already confirmed in diseases such as acute diarrhoea, antibiotic- associated diarrhoea, functional gastrointestinal disorders, inflammatory bowel disease, etc[28]. H. pylori infection is also a field in which particular interest in using probiotics is arising.

GASTRIC MICROBIOTA

Lactobacillus species are acid resistant and they are normally present in healthy gastric microbiota. Some strains can even adhere to gastric epithelial cells so they can stay longer than other bacteria in the stomach[29]. These has been confirmed in a study where it was possible to detect L. reuteri 55730 adhering to gastric epithelial cells of healthy volunteers a few hours after oral administration[30]. Elliott et al[31] has observed that in a rat with active gastric ulcer the local microbiota changed toward Gram-negative bacteria including E. coli while the Lactobacullus species almost disappeared. In the process of gastric mucosa healing the Lactobacillus population reappeared again and these process was accelerated by the oral lactulose administration. This study suggests that normal gastric microbiota participate in maintaining healthy mucosa. In addition, the oral intake of probiotics may reinforce protective functions in the stomach.

DIFFERENT MECHANISMS OF PROBIOTICS ANTIBACTERIAL ACTION

Probiotics can inhibit H. pylori by several immunological and non-immunological mechanisms.

Probiotics are capable of modifying immunologic response of the host. Neutrophils, lymphocytes, plasma cells, and macrophages are involved in the inflammatory response to H. pylori. The consequences are increased levels of pro-inflammatory cytokines, such as IL-1β, IL-2, IL-6, IL-8 and tumor necrosis factor α in the gastric mucosa[32]. Probiotics like L. salivarius WB 1004 have in vitro reduced IL-8 secretion by gastric epithelial cells[33]. It looks like that at least in vitro L. acidophilus can improve H. pylori-induced gastric inflammation by inactivating the Smad7 and NFκB pathways[34]. Furthermore, Zhou et al[35] demonstrated that L. bulgaricus inhibited the activation of the TLR4 signaling pathway and IL-8 production induced by H. pylori lipopolysaccharide in SGC-7901 cells. The gastric inflammation can be controlled to some level by different strains of Lactobacilli through inactivating JAK2 through JAK– STAT pathways and through higher expression of SOCS protein family[36]. H. pylori also induce humoral response of gastric mucosa, which may contribute to gastric mucosal damage. Lactobacilli were able to increase the local IgA concentration and decrease specific anti H. pylori IgG antibodies in animal models[37,38].

Among non-immunological mechanisms probiotics are capable to influence on bacterial growth by secreting antibacterial substances such as lactic acid, short chain fatty acids, hydrogen peroxide and bactericins. The metabolites can diminish the number of spiral bacteria[39]. Lactic acid has probably an additional effect on H. pylori by lowering the pH and inhibiting the urease[40]. L. acidophilus CRL 639 secret an autolysin, a proteinaceous compound released after cell lysis, which has some antibacterial activity[41]. Substances, similar to isocoumarin antibiotics, are produced by B. subtilis and those can also kill H. pylori bacteria[42]. L. reuteri ATCC 55730 produce unique substance called reuterina which suppress growth of spiral bacteria[43].

Mucins are high-molecular-weight glycoproteins that protect the gastric epithelium. The gastric surface-type mucin is decreased in infected patients, because H. pylori suppresses the expression of MUC5AC and MUC1 genes[44]. It has been shown that L. plantarum strain 299v increases the expression of MUC2 gene and that L. rhamnosus GG stimulates the expression of MUC3 gene and the subsequent extracellular secretion of mucin by colon cell cultures[45,46]. Therefore, the ability of some probiotic strains to increase mucin production can protect the gastric mucosal barrier against the adherence of pathogenic bacteria such as H. pylori.

Adherence of H. pylori to the host tissue is of vital importance for colonization the gastric mucosa[47]. There are several proposed mechanisms of anti-adherence activity of probiotics. L. reuteri and W. confusa are capable of competing with H. pylori strains for adhesion sites. For example, L. reuteri strains, JCM 1081 and TM 105, inhibit the binding of spiral bacteria to glycolipid receptors asialo-GMI and sulfatide[48]. In addition, Sakarya et al[49] demonstrated that S. boulardii contains neuraminidase activity, which removes surface α(2-3)-linked sialic acid, the ligand for the sialic acid-binding H. pylori adhesion.

POSSIBLE ROLE OF PROBIOTICS IN H. PYLORI TREATMENT

Eradication failure rate of more than 20% and high percentage of adverse effects of antibiotic therapy are main problems in standard eradication therapy protocols. Furthermore, lover compliance due to adverse events results in higher antibiotic resistance of bacterial strains. Studies published to date suggest that probiotics can have dual role in fighting against H. pylori infection. They diminish the frequency of gastrointestinal adverse events caused by antibiotic therapy and increase the eradication rate. The probiotics have been experimentally used as single therapy in eradication protocols or as therapeutic agent used concomitantly with standard eradication therapy.

Probiotics as single therapy in H. pylori treatment

The first in vitro positive results were published in 1989[50]. Bhatia et al[50] discovered that H. pylori growth in vitro was inhibited, if L. acidophilus was present in the culture. Michetti et al[51] studied for the first time the effect of probiotics [L. acidophilus (La1)] on the H. pylori colonisation in humans. The study showed that the density of bacterial load diminishes in the probiotic group of asymptomatic patients, whereas complete eradication of H. pylori was not successful. Similarly, Wang et al[52] revealed that after intake of B. lactis Bb12 and L. acidophilus La5 in H. pylori infected adults a decrease in urea breath test values was detected. The bacterial load was evaluated by the semi-quantitative 13C-urea breath test in subjects treated with L. johnsonii La1[53], L. brevis CD2 lyophilized bacteria[54], B. bifidum BF-1[55], L. reuteri ATCC 55730[43], L. gasseri OLL 2716[56], and with multispecies probiotics such as a combination of L. reuteri DSM 17938 and L. reuteri ATCC PTA 6457[57] or combination of L. rhamnosus GG, L. rhamnosus LC705, P. Freudenreichii JS and B. lactis Bb12[58]. Regardless of the used probiotics the authors reported significant decrease in 13C-urea breath test values in the probiotic group of studied patients.

Gotteland et al[59] included 182 asymptomatic children infected with H. pylori and they were divided into four groups: standard triple therapy group, S. boulardii and inulin synbiotic group, L. acidophilus LB probiotic group, or control group without any therapy. Statistically significant decreases of urea breath test values were detected in two groups: the standard triple group and the S. boulardii inulin synbiotic group of children. The authors concluded that S. boulardii can lower the bacterial load in the gastric mucosa of children infected with H. pylori bacteria. Surprisingly, in 12% of children in the second group treated with S. boulardii and inulin H. pylori even the eradication was successful. The same author carried out randomized, double-blind study in 295 asymptomatic children infected with H. pylori[60]. The study compared eradication rates after 3 wk of therapy with (1) placebo juice/L. johnsonii La1; (2) cranberry juice /L. johnsonii La1; (3) placebo juice/heat-killed L. johnsonii La1; and (4) cranberry juice/heat-killed L. johnsonii La1. Except for the placebo group the eradication rates were above 14% in all tested groups, but didn’t statistically significantly differ between each other. Cruchet et al[61] included 326 infected children in a similar study. The children were divided into five groups. They were treated for one month either with live or heat-killed L. johnsonii La1 or either with live or inactivated L. paracasei ST11. The fifth group was the control group. Statistically significant changes in 13C-urea breath test were observed only in the group of pediatric patients treated with live L. johnsonii La1 probiotics.

In conclusion, there are only a few studies evaluating the effect of probiotics as monotherapy on H. pylori eradication rate. The results of the studies suggest that specific probiotics, such as S. boulardii and L. johnsonni La1 probably diminish the bacterial load, but not completely eradicate the H. pylori bacteria.

Probiotics as adjuvant therapy in standard eradication protocols

Several systematic reviews and meta-analyses regarding effect of probiotics as adjuvant therapy to standard treatment of H. pylori infection have been published[62,63]. The authors suggested that probiotics supplementation in general probably increase the eradication rate and reduce the frequency of adverse effects due to double antibiotic therapy. However, the beneficial effects of probiotics seem to be strain-specific, thus, collecting data on different strains in meta-analysis may result in misleading conclusions. Regarding this, a better approach is to pool the data on single probiotics strain and perform a meta-analysis. Szajewska et al[64] recently published a systematic review to evaluate the effects of supplementations with S. boulardii to standard triple therapy protocol on H. pylori eradication rate. Five randomized controlled trials of good methodological quality involving 1307 patients were identified. Among them only 90 children were included. The daily dose of S. boulardii ranged from 500 mg to 1000 mg and the duration of the therapy was from 2-4 wk. From four trials the complete data on the eradication rates were available. In 80% of the included patients treated with S. boulardii along with triple therapy the eradication was confirmed by standard diagnostic tools. In the control group 9% lover absolute eradication rate was detected (71%, 324 of the 455 patients). The authors concluded that compared with placebo or no intervention, S. boulardii given along with triple therapy significantly increased the eradication rate [relative risk (RR) = 1.13, 95%CI: 1.05-1.21]. The secondary endpoints of the same meta-analysis were also to determine the effect of S. boulardii on therapy-related adverse effects. About 24.3% of patients experienced adverse effects in control group treated with triple therapy, compared to 12.9% of patients in probiotic group. Thus, the significant difference was found between the S. boulardi group and the control group with respect to the risk of overall adverse effects (five randomised control trials, n = 1305, RR = 0.46, 95%CI: 0.3-0.7). The authors analyse also the data of specific adverse effects. With regard to epigastric pain, taste disturbance/dry mouth, nausea or abdominal gas/bloating no significant difference was found between the studied groups. On the other hand, the risk of therapy related diarrhoea was statistically significantly lower in the probiotic group compared with the control group treated only with antibiotics and proton pump inhibitor (5.6% vs 12.2%, RR = 0.47, 95%CI: 0.32-0.69). The conclusions of the meta-analysis were that the concomitant use of S. boulardii with triple therapy moderately increases H. pylori eradication rates and decreases antibiotic related adverse effects, especially diarrhoea.

However, more recent studies published by Song et al[65] and Zojaji et al[66] didn’t confirm positive impact of S. boulardii on eradication rate. In Song’s study 991 H. pylori infected patients were recruited. Patients in group A were treated only with two antibiotics and proton pump inhibitor, in group B S. boulardii was added for one month, and in group C the same regimen was used and in addition mucoprotective agent DA-9601 derived from Artemisia asiatica was concomitantly prescribed. Interestingly, the eradication rate was significantly higher in group B and C compared to group A only if intention to treat analysis was performed (P = 0.003), whereas the eradication rate difference of per protocol patients analysis was not significant. The conclusion was that supplementation with S. boulardi could be effective for improving eradication rates by reducing adverse effects thus helping completion of eradication therapy. Zojaji et al[66] included 160 adult patients. In the study protocol the probiotic S. boulardii was added to clarithromycin, amoxicillin and omeprazole for two weeks. The study showed that probiotics decrease the frequency of adverse events due to antibiotic therapy, but didn’t increase the eradication rate of H. pylori.

Tong et al[62] systematically evaluated the effectiveness of supplementation with different probiotics in increasing H. pylori eradication rates. In the publication they also made sub-analysis for different probiotics preparations. In eight of fourteen reported randomized trials single probiotics strain was used. In tree trials, Lactobacillus species was administered to the standard eradication therapy[67-69]. However, increased H. pylori eradication rate in Lactobacillus group was reported in only two studies published by Sýkora et al[69] and Canducci et al[67]. Overall in the tree studies the eradication rates were 70% in control group and 84% in probiotic supplemented group, which is a statistically significant difference (RR = 2.09, 95%CI: 1.28-3.41). In addition, this meta-analysis revealed that adding probiotics to standard eradication protocols reduces adverse effects during treatment (25% vs 39%, RR = 0.44, 95%CI: 0.30-0.66). The positive impact on diarrhoea and taste disturbance was most prominent.

The effectiveness of L. GG in children with H. pylori infection was studied in a trial from Poland[70]. Of the 83 children tested, 34 children in a probiotics group received L. GG 109 twice daily for one week concomitantly with triple therapy. No significant difference in H. pylori eradication rates between the probiotic and the control group were found (RR = 0.98, 95%CI: 0.7-1.4). This result is in accordance with trials in adult population. Armuzzi et al[68] included 60 infected asymptomatic patients in a prospective study in which patients were treated with rabeprazole, clarithromycin tinidazole and L. GG or with the same triple therapy and placebo. Diarrhoea, nausea and taste disturbance were significantly reduced in the L. GG supplemented group (relative risk = 0.1, 95%CI: 0.1 ± 0.9; relative risk = 0.3, 95%CI: 0.1 ± 0.9; relative risk = 0.5, 95%CI: 0.2 ± 0.9, respectively). In another Italian study 85 asymptomatic adults were included, and the conclusion of the study was that L. GG supplementation to standard therapy beneficially affects treatment related adverse effects[71]. However, it seems that L. GG has no effect on eradication rates.

Demonstration that L. reuteri ATCC 55730 is able to colonize the stomach and duodenum prompted studies regarding the effect of this strain on H. pylori eradication rates and the frequency of side effects[30]. In a recent study conducted in Italy, Ojetti et al[72] recruited 90 patients in their study. L. reuteri supplementation was concomitantly used for 14 d with second line therapy receiving esomeprazole, levofloxacin and amoxicillin in patients infected with H. pylori. Probiotic supplementation increased the eradication rate in treated patients (group 1: 36/45, 80%; group 2: 28/45 62%; P < 0.05). In addition, the incidence of side effects associated with antibiotic therapy was also significantly lower in the probiotic group. This is in agreement with a trial that also evaluated the impact of L. reuteri ATCC 55730 (108 CFU for twenty days) as an adjuvant to 10-d sequential therapy in a group of 40 H. pylori infected children[73]. The Gastrointestinal Symptom Rating Score was lower in the group of children treated with L. reuteri (3.2 vs 5.8, P < 0.009). However, the use of L. reuteri as an adjunct to the sequential eradication therapy had no effect on eradication rates (17/20 vs 16/20). In 2009, Francavilla et al[43] compared the eradication rates and antibiotic adverse effects in 40 H. pylori positive subjects who were receiving for a month placebo or L. reuteri (108 CFU) once a day. At the end of the trial patients received standard 10-d sequential eradication therapy. Four week supplementation with L. reuteri was effective in lowering gastrointestinal adverse effects and also in reducing bacterial load, whereas no statistical significant difference in eradication rates was observed (88% vs 82%). Recently, the same group published results of the double-blind placebo-controlled randomized study using a combination of two L. reuteri strains (L. reuteri DSM 17938 and L. reuteri ATCC 55730) as an adjunct to triple eradication therapy[57]. L. reuteri DSM 17938 is a safe daughter strain of previously used L. reuteri ATCC 55730 in which two plasmids coding antibiotic resistance were removed. The new L. reuteri ATCC 55730 strain seems to have strong anti-inflammatory properties. A combination of two L. reuteri strains or placebo was given for two months concomitantly with one-week triple eradication therapy in a second part of the study. A significant reduction of adverse effects was shown in the group treated with eradication therapy and the combination of two L. reuteri strains, whereas H. pylori eradication rate was only slightly but not significantly increased in the same group of patients. In another study published by Emara et al[74] triple therapy supplemented with L. reuteri increased eradication rate by 8.6% and reduced the frequency of side effects. L. reuteri has a positive influence on gastrointestinal side effects especially diarrhoea, but conflicting reports regarding the impact of L. reuteri on H. pylori eradication rates demand further studies, especially because new strain combination was developed.

The use of commercial yogurt containing B. animalis and L. casei combined with conventional triple therapy was investigated in two studies. Sheu et al[75] tested the efficacy of this yogurt as adjuvant to triple treatment. The difference in eradication rates of H. pylori infection in two groups was statistically significant in favour of probiotic group (73/80 vs 63/80, P < 0.05). Only patients supplemented with yogurt showed restoration of the percentage of Bifidobacterium in the stools at the end of the study to the level in the stools on enrolment. Goldman et al[76] was not able to confirm the important role of yogurt supplementation to standard triple therapy in a pediatric study. Contrary to Sheu study protocol, in which yogurt was administrated for five weeks, they continued with the probiotics treatment for three months. In spite of prolonged therapy they found no difference in H. pylori eradication rates among the yogurt and the placebo groups of patients (14/33 vs 13/32, P value is not significant).

Effect of pre-treatment with L. gasseri OLL2716 on first-line eradication therapy was studied in a trial published by Deguchi et al[77]. About 229 infected patients were randomized into two groups, either one-week triple therapy (rabeprazole, clarithromycin, amoxicillin) or the same therapy with addition of probiotics. Overall eradication was significantly better in the studied probiotic group [intention to treat (P = 0.018)/per protocol (P = 0.041)], but more prospective studies are needed to evaluate the role of L. gasseri OLL2716 in the treatment in H. pylori infection.

Beneficial effects of adding fermented milk product containing L. casei DN-114 001 to the triple therapy on the eradication rate of H. pylori infection in children were showed in a multicentre study from the Check Republic[67]. Eighty-six symptomatic children infected with H. pylori were randomized either to receive the omeprazole, amoxicillin, clarithromycin or the same regimen with addition of probiotic for 7 d. Eradication rate was statistically significantly higher in the probiotic group (P = 0.0045). B. claussi have also been studied as a possible adjunct to standard therapy for H. pylori eradication, but the positive effect on eradication rate was not confirmed[78]. The characteristics of important studies where probiotics were used as adjuvant therapy to eradication treatment are presented in Table 1.

Table 1 Probiotics as adjuvant therapy in Helicobacter pylori eradication protocols.
Ref.Therapy
Study
Eradication rate
Adverse effects
Eradication therapy (d)Probiotic therapy(d)No. ptsReass. ofH. pyloriH. pylori + probiotic therapyH. pylori therapyH. pylori + probiotic therapyH. pylori therapy
Cindoruk et al[79]PPI + AC (14)S. boulardii (14)124UBT44/62137/6214/62137/62
Cremonini et al[71]PPI + CT (7)S. boulardii (14)43UBT17/2016/203/21112/20
Duman et al[80]PPI + AC (14)S. boulardii (14)389---17/204131/185
Hurduc et al[81]PPI + AC (10)S. boulardii (28)90UBT, hist.45/4834/424/48113/42
Song et al[65]PPI + AC (7)S. boulardii (28)661UBT264/330237/33148/330163/331
Zojaji et al[66]PPI + AC (14)S. boulardii (14)160UBT75/8065/8014/80129/80
Canducci et al[67]PPI + AC (7)L. acidophilus (7 )120UBT42/60152/60--
Sýkora et al[69]PPI + AC (7)L. casei (14)86UBT33/39127/477/399/47
Armuzzi et al[68]PPI + CT (7)L. GG (14)60UBT25/3024/3012/30120/30
Szajewska et al[70]PPI + AC (7)L. GG (14)83UBT, hist., RUT23/3422/3218/3513/32
Cremonini et al[71]PPI + CT (7)L. GG (14)85UBT16/2116/203/20112/20
Ojetti et al[72]PPI + ALL. reuteri (14 )90UBT36/45127/4510/45126/45
Lionetti et al[73]PPI + ACT (10)L. reuteri (20 )42UBT17/2016/203/2019/20
Francavilla et al[57]PPI + (7)L. reuteri (2 strains) (60)100UBT
Emara et al[74]PPI + AC (14)L. reuteri (28)70hist., RUT26/3523/3510/35124/35
Sheu et al[75]PPI + AC (7)B. animalis, L. casei (35)160hist., RUT, UBT73/80163/8015/80153/80
Goldman et al[76]PPI + AC (7)B. animalis, L. casei (90)65UBT14/3313/32--
Deguchi et al[77]PPI + AC (7)L. gasseri (28)229UBT95/111179/106--
Nista et al[78]PPI + AC (7)B. clausii (14)120UBT39/5037/5021/50130/50
CONCLUSION

Chronic infection with H. pylori is a well-described risk factor for ulcer disease, gastric adenocarcinoma and MALT lymphoma. Therefore, eradication of H. pylori is a primary goal in symptomatic patients. The eradication rates achieved by classic triple therapy consisted from proton pump inhibitor and double antibiotic therapy are quite low and range from 60% to 80%, due to resistance to antibiotics and to moderate patient compliance. Antibiotic-associated gastrointestinal adverse events are the major cause for lower compliance. Therefore, probiotics were proposed as a useful adjunct to improve eradication rate and to decrease the frequency of adverse effects.

So far, mostly different types of Lactobacillus and S. boulardii were tested. The above-mentioned probiotics most probably decrease the bacterial load but don’t eradicate H. pylori completely in the gastric mucosa, if they are used as monotherapy. On the contrary, some probiotics when added to classical triple therapy may increase eradication rates. A reasonable amount of evidence exists that supplementation with S. boulardii is a useful concomitant therapy in the standard H. pylori eradication treatment protocol and most probably increases the eradication rate. L. reuteri is also a good candidate for adjunctive therapy, but more positive studies are needed. The effect of other probiotics strains is less well described.

Side effects, caused by double antibiotic therapy, can be lowered with probiotics. Probiotic strains, such as S. boulardii, L. reuteri and L. GG, decrease gastrointestinal antibiotic associated adverse effects, especially diarrhoea. The Maastricht IV/Florence Consensus Report suggests that certain probiotics and prebiotics show promising results as an adjuvant treatment in reducing side effects[13].

Footnotes

P- Reviewer: D'Elios MM, Eren B, Malnick S, Sakarya S S- Editor: Yu J L- Editor: A E- Editor: Wang CH

References
1.  Solnick JV, Schauer DB. Emergence of diverse Helicobacter species in the pathogenesis of gastric and enterohepatic diseases. Clin Microbiol Rev. 2001;14:59-97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 273]  [Cited by in F6Publishing: 280]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
2.  Josenhans C, Suerbaum S. The role of motility as a virulence factor in bacteria. Int J Med Microbiol. 2002;291:605-614.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 442]  [Cited by in F6Publishing: 436]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
3.  Weeks DL, Eskandari S, Scott DR, Sachs G. A H+-gated urea channel: the link between Helicobacter pylori urease and gastric colonization. Science. 2000;287:482-485.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 364]  [Cited by in F6Publishing: 312]  [Article Influence: 13.0]  [Reference Citation Analysis (1)]
4.  D’Elios MM, Czinn SJ. Immunity, inflammation, and vaccines for Helicobacter pylori. Helicobacter. 2014;19 Suppl 1:19-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 37]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
5.  Osaki T, Yamaguchi H, Taguchi H, Fukuda M, Kawakami H, Hirano H, Watanabe S, Takagi A, Kamiya S. Establishment and characterisation of a monoclonal antibody to inhibit adhesion of Helicobacter pylori to gastric epithelial cells. J Med Microbiol. 1998;47:505-512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 29]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
6.  Sakamoto S, Watanabe T, Tokumaru T, Takagi H, Nakazato H, Lloyd KO. Expression of Lewisa, Lewisb, Lewisx, Lewisy, siayl-Lewisa, and sialyl-Lewisx blood group antigens in human gastric carcinoma and in normal gastric tissue. Cancer Res. 1989;49:745-752.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Yamaoka Y. Increasing evidence of the role of Helicobacter pylori SabA in the pathogenesis of gastroduodenal disease. J Infect Dev Ctries. 2008;2:174-181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 50]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
8.  Walz A, Odenbreit S, Stühler K, Wattenberg A, Meyer HE, Mahdavi J, Borén T, Ruhl S. Identification of glycoprotein receptors within the human salivary proteome for the lectin-like BabA and SabA adhesins of Helicobacter pylori by fluorescence-based 2-D bacterial overlay. Proteomics. 2009;9:1582-1592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 35]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
9.  Van de Bovenkamp JH, Mahdavi J, Korteland-Van Male AM, Büller HA, Einerhand AW, Borén T, Dekker J. The MUC5AC glycoprotein is the primary receptor for Helicobacter pylori in the human stomach. Helicobacter. 2003;8:521-532.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Kawakami E, Machado RS, Ogata SK, Langner M. Decrease in prevalence of Helicobacter pylori infection during a 10-year period in Brazilian children. Arq Gastroenterol. 2008;45:147-151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
11.  Elitsur Y, Dementieva Y, Rewalt M, Lawrence Z. Helicobacter pylori infection rate decreases in symptomatic children: a retrospective analysis of 13 years (1993-2005) from a gastroenterology clinic in West Virginia. J Clin Gastroenterol. 2009;43:147-151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 30]  [Article Influence: 2.0]  [Reference Citation Analysis (1)]
12.  Parsonnet J, Hansen S, Rodriguez L, Gelb AB, Warnke RA, Jellum E, Orentreich N, Vogelman JH, Friedman GD. Helicobacter pylori infection and gastric lymphoma. N Engl J Med. 1994;330:1267-1271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1287]  [Cited by in F6Publishing: 1204]  [Article Influence: 40.1]  [Reference Citation Analysis (0)]
13.  Malfertheiner P, Megraud F, O’Morain CA, Atherton J, Axon AT, Bazzoli F, Gensini GF, Gisbert JP, Graham DY, Rokkas T. Management of Helicobacter pylori infection--the Maastricht IV/ Florence Consensus Report. Gut. 2012;61:646-664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1541]  [Cited by in F6Publishing: 1494]  [Article Influence: 124.5]  [Reference Citation Analysis (3)]
14.  Homan M, Luzar B, Kocjan BJ, Orel R, Mocilnik T, Shrestha M, Kveder M, Poljak M. Prevalence and clinical relevance of cagA, vacA, and iceA genotypes of Helicobacter pylori isolated from Slovenian children. J Pediatr Gastroenterol Nutr. 2009;49:289-296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
15.  Homan M, Šterbenc A, Kocjan BJ, Luzar B, Zidar N, Orel R, Poljak M. Prevalence of the Helicobacter pylori babA2 gene and correlation with the degree of gastritis in infected Slovenian children. Antonie Van Leeuwenhoek. 2014;106:637-645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 10]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
16.  Kodama M, Murakami K, Okimoto T, Abe T, Nakagawa Y, Mizukami K, Uchida M, Inoue K, Fujioka T. Helicobacter pylori eradication improves gastric atrophy and intestinal metaplasia in long-term observation. Digestion. 2012;85:126-130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 46]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
17.  Taube C, Müller A. The role of Helicobacter pylori infection in the development of allergic asthma. Expert Rev Respir Med. 2012;6:441-449.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
18.  Wang Q, Yu C, Sun Y. The association between asthma and Helicobacter pylori: a meta-analysis. Helicobacter. 2013;18:41-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 62]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
19.  Vo HD, Goli S, Gill R, Anderson V, Stefanov DG, Xu J, Kulsum-Mecci N, Schwarz SM, Rabinowitz SS. Inverse correlation between Helicobacter pylori colonization and obesity in a cohort of inner city children. Helicobacter. 2015;20:64-68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
20.  Qian B, Ma S, Shang L, Qian J, Zhang G. Effects of Helicobacter pylori eradication on gastroesophageal reflux disease. Helicobacter. 2011;16:255-265.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 46]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
21.  Mégraud F. H pylori antibiotic resistance: prevalence, importance, and advances in testing. Gut. 2004;53:1374-1384.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 607]  [Cited by in F6Publishing: 648]  [Article Influence: 32.4]  [Reference Citation Analysis (1)]
22.  Saracino IM, Zullo A, Holton J, Castelli V, Fiorini G, Zaccaro C, Ridola L, Ricci C, Gatta L, Vaira D. High prevalence of primary antibiotic resistance in Helicobacter pylori isolates in Italy. J Gastrointestin Liver Dis. 2012;21:363-365.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Sasaki M, Ogasawara N, Utsumi K, Kawamura N, Kamiya T, Kataoka H, Tanida S, Mizoshita T, Kasugai K, Joh T. Changes in 12-Year First-Line Eradication Rate of Helicobacter pylori Based on Triple Therapy with Proton Pump Inhibitor, Amoxicillin and Clarithromycin. J Clin Biochem Nutr. 2010;47:53-58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 73]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
24.  Misiewicz JJ. Management of Helicobacter pylori-related disorders. Eur J Gastroenterol Hepatol. 1997;9 Suppl 1:S17-20; discussion S20-1, S27-9.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Henry A, Batey RG. Enhancing compliance not a prerequisite for effective eradication of Helicobacter pylori: the HelP Study. Am J Gastroenterol. 1999;94:811-815.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 33]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
26.  Food and Agriculture Organisation of the United Nations; World Health Organisation. Guidelines for the evaluation of probiotics in food: joint FAO/WHO Working Group report on drafting guidelines for the evaluation of probiotics in food.  Available from: ftp://ftp.fao.org/es/esn/food/wgreport2.pdf.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Sullivan A, Nord CE. Probiotics and gastrointestinal diseases. J Intern Med. 2005;257:78-92.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 106]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
28.  Gill HS, Guarner F. Probiotics and human health: a clinical perspective. Postgrad Med J. 2004;80:516-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 195]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
29.  Sherman LA, Savage DC. Lipoteichoic acids in Lactobacillus strains that colonize the mouse gastric epithelium. Appl Environ Microbiol. 1986;52:302-304.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Valeur N, Engel P, Carbajal N, Connolly E, Ladefoged K. Colonization and immunomodulation by Lactobacillus reuteri ATCC 55730 in the human gastrointestinal tract. Appl Environ Microbiol. 2004;70:1176-1181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 245]  [Cited by in F6Publishing: 236]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
31.  Elliott SN, Buret A, McKnight W, Miller MJ, Wallace JL. Bacteria rapidly colonize and modulate healing of gastric ulcers in rats. Am J Physiol. 1998;275:G425-G432.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Goodwin CS, Mendall MM, Northfield TC. Helicobacter pylori infection. Lancet. 1997;349:265-269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 142]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
33.  Borruel N, Casellas F, Antolín M, Llopis M, Carol M, Espíin E, Naval J, Guarner F, Malagelada JR. Effects of nonpathogenic bacteria on cytokine secretion by human intestinal mucosa. Am J Gastroenterol. 2003;98:865-870.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 108]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
34.  Yang YJ, Chuang CC, Yang HB, Lu CC, Sheu BS. Lactobacillus acidophilus ameliorates H. pylori-induced gastric inflammation by inactivating the Smad7 and NFκB pathways. BMC Microbiol. 2012;12:38.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 89]  [Article Influence: 7.4]  [Reference Citation Analysis (1)]
35.  Zhou C, Ma FZ, Deng XJ, Yuan H, Ma HS. Lactobacilli inhibit interleukin-8 production induced by Helicobacter pylori lipopolysaccharide-activated Toll-like receptor 4. World J Gastroenterol. 2008;14:5090-5095.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
36.  Lee JS, Paek NS, Kwon OS, Hahm KB. Anti-inflammatory actions of probiotics through activating suppressor of cytokine signaling (SOCS) expression and signaling in Helicobacter pylori infection: a novel mechanism. J Gastroenterol Hepatol. 2010;25:194-202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 57]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
37.  Vitiñi E, Alvarez S, Medina M, Medici M, de Budeguer MV, Perdigón G. Gut mucosal immunostimulation by lactic acid bacteria. Biocell. 2000;24:223-232.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Sgouras D, Maragkoudakis P, Petraki K, Martinez-Gonzalez B, Eriotou E, Michopoulos S, Kalantzopoulos G, Tsakalidou E, Mentis A. In vitro and in vivo inhibition of Helicobacter pylori by Lactobacillus casei strain Shirota. Appl Environ Microbiol. 2004;70:518-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 212]  [Cited by in F6Publishing: 223]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
39.  Midolo PD, Lambert JR, Hull R, Luo F, Grayson ML. In vitro inhibition of Helicobacter pylori NCTC 11637 by organic acids and lactic acid bacteria. J Appl Bacteriol. 1995;79:475-479.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Lesbros-Pantoflickova D, Corthésy-Theulaz I, Blum AL. Helicobacter pylori and probiotics. J Nutr. 2007;137:812S-818S.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Lorca GL, Wadström T, Valdez GF, Ljungh A. Lactobacillus acidophilus autolysins inhibit Helicobacter pylori in vitro. Curr Microbiol. 2001;42:39-44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 100]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
42.  Pinchuk IV, Bressollier P, Verneuil B, Fenet B, Sorokulova IB, Mégraud F, Urdaci MC. In vitro anti-Helicobacter pylori activity of the probiotic strain Bacillus subtilis 3 is due to secretion of antibiotics. Antimicrob Agents Chemother. 2001;45:3156-3161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 140]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
43.  Francavilla R, Lionetti E, Castellaneta SP, Magistà AM, Maurogiovanni G, Bucci N, De Canio A, Indrio F, Cavallo L, Ierardi E. Inhibition of Helicobacter pylori infection in humans by Lactobacillus reuteri ATCC 55730 and effect on eradication therapy: a pilot study. Helicobacter. 2008;13:127-134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 102]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
44.  Byrd JC, Yunker CK, Xu QS, Sternberg LR, Bresalier RS. Inhibition of gastric mucin synthesis by Helicobacter pylori. Gastroenterology. 2000;118:1072-1079.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 109]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
45.  Mack DR, Michail S, Wei S, McDougall L, Hollingsworth MA. Probiotics inhibit enteropathogenic E. coli adherence in vitro by inducing intestinal mucin gene expression. Am J Physiol. 1999;276:G941-G950.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Mack DR, Ahrne S, Hyde L, Wei S, Hollingsworth MA. Extracellular MUC3 mucin secretion follows adherence of Lactobacillus strains to intestinal epithelial cells in vitro. Gut. 2003;52:827-833.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 434]  [Cited by in F6Publishing: 395]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
47.  Guruge JL, Falk PG, Lorenz RG, Dans M, Wirth HP, Blaser MJ, Berg DE, Gordon JI. Epithelial attachment alters the outcome of Helicobacter pylori infection. Proc Natl Acad Sci USA. 1998;95:3925-3930.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 226]  [Cited by in F6Publishing: 238]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
48.  Mukai T, Asasaka T, Sato E, Mori K, Matsumoto M, Ohori H. Inhibition of binding of Helicobacter pylori to the glycolipid receptors by probiotic Lactobacillus reuteri. FEMS Immunol Med Microbiol. 2002;32:105-110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
49.  Sakarya S, Gunay N. Saccharomyces boulardii expresses neuraminidase activity selective for α2,3-linked sialic acid that decreases Helicobacter pylori adhesion to host cells. APMIS. 2014;122:941-950.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 50]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
50.  Bhatia SJ, Kochar N, Abraham P, Nair NG, Mehta AP. Lactobacillus acidophilus inhibits growth of Campylobacter pylori in vitro. J Clin Microbiol. 1989;27:2328-2330.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Michetti P, Dorta G, Wiesel PH, Brassart D, Verdu E, Herranz M, Felley C, Porta N, Rouvet M, Blum AL. Effect of whey-based culture supernatant of Lactobacillus acidophilus (johnsonii) La1 on Helicobacter pylori infection in humans. Digestion. 1999;60:203-209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 222]  [Cited by in F6Publishing: 226]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
52.  Wang KY, Li SN, Liu CS, Perng DS, Su YC, Wu DC, Jan CM, Lai CH, Wang TN, Wang WM. Effects of ingesting Lactobacillus- and Bifidobacterium-containing yogurt in subjects with colonized Helicobacter pylori. Am J Clin Nutr. 2004;80:737-741.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Gotteland M, Cruchet S. Suppressive effect of frequent ingestion of Lactobacillus johnsonii La1 on Helicobacter pylori colonization in asymptomatic volunteers. J Antimicrob Chemother. 2003;51:1317-1319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 58]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
54.  Linsalata M, Russo F, Berloco P, Caruso ML, Matteo GD, Cifone MG, Simone CD, Ierardi E, Di Leo A. The influence of Lactobacillus brevis on ornithine decarboxylase activity and polyamine profiles in Helicobacter pylori-infected gastric mucosa. Helicobacter. 2004;9:165-172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 66]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
55.  Miki K, Urita Y, Ishikawa F, Iino T, Shibahara-Sone H, Akahoshi R, Mizusawa S, Nose A, Nozaki D, Hirano K. Effect of Bifidobacterium bifidum fermented milk on Helicobacter pylori and serum pepsinogen levels in humans. J Dairy Sci. 2007;90:2630-2640.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 42]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
56.  Sakamoto I, Igarashi M, Kimura K, Takagi A, Miwa T, Koga Y. Suppressive effect of Lactobacillus gasseri OLL 2716 (LG21) on Helicobacter pylori infection in humans. J Antimicrob Chemother. 2001;47:709-710.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 222]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
57.  Francavilla R, Polimeno L, Demichina A, Maurogiovanni G, Principi B, Scaccianoce G, Ierardi E, Russo F, Riezzo G, Di Leo A. Lactobacillus reuteri strain combination in Helicobacter pylori infection: a randomized, double-blind, placebo-controlled study. J Clin Gastroenterol. 2014;48:407-413.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 71]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
58.  Myllyluoma E, Kajander K, Mikkola H, Kyrönpalo S, Rasmussen M, Kankuri E, Sipponen P, Vapaatalo H, Korpela R. Probiotic intervention decreases serum gastrin-17 in Helicobacter pylori infection. Dig Liver Dis. 2007;39:516-523.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 30]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
59.  Gotteland M, Poliak L, Cruchet S, Brunser O. Effect of regular ingestion of Saccharomyces boulardii plus inulin or Lactobacillus acidophilus LB in children colonized by Helicobacter pylori. Acta Paediatr. 2005;94:1747-1751.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 80]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
60.  Gotteland M, Andrews M, Toledo M, Muñoz L, Caceres P, Anziani A, Wittig E, Speisky H, Salazar G. Modulation of Helicobacter pylori colonization with cranberry juice and Lactobacillus johnsonii La1 in children. Nutrition. 2008;24:421-426.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Cruchet S, Obregon MC, Salazar G, Diaz E, Gotteland M. Effect of the ingestion of a dietary product containing Lactobacillus johnsonii La1 on Helicobacter pylori colonization in children. Nutrition. 2003;19:716-721.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 123]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
62.  Tong JL, Ran ZH, Shen J, Zhang CX, Xiao SD. Meta-analysis: the effect of supplementation with probiotics on eradication rates and adverse events during Helicobacter pylori eradication therapy. Aliment Pharmacol Ther. 2007;25:155-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 158]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
63.  Zhu R, Chen K, Zheng YY, Zhang HW, Wang JS, Xia YJ, Dai WQ, Wang F, Shen M, Cheng P. Meta-analysis of the efficacy of probiotics in Helicobacter pylori eradication therapy. World J Gastroenterol. 2014;20:18013-18021.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Szajewska H, Horvath A, Piwowarczyk A. Meta-analysis: the effects of Saccharomyces boulardii supplementation on Helicobacter pylori eradication rates and side effects during treatment. Aliment Pharmacol Ther. 2010;32:1069-1079.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 153]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
65.  Song MJ, Park DI, Park JH, Kim HJ, Cho YK, Sohn CI, Jeon WK, Kim BI. The effect of probiotics and mucoprotective agents on PPI-based triple therapy for eradication of Helicobacter pylori. Helicobacter. 2010;15:206-213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 65]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
66.  Zojaji H, Ghobakhlou M, Rajabalinia H, Ataei E, Jahani Sherafat S, Moghimi-Dehkordi B, Bahreiny R. The efficacy and safety of adding the probiotic Saccharomyces boulardiito standard triple therapy for eradication of H.pylori: a randomized controlled trial. Gastroenterol Hepatol Bed Bench. 2013;6:S99-S104.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Canducci F, Armuzzi A, Cremonini F, Cammarota G, Bartolozzi F, Pola P, Gasbarrini G, Gasbarrini A. A lyophilized and inactivated culture of Lactobacillus acidophilus increases Helicobacter pylori eradication rates. Aliment Pharmacol Ther. 2000;14:1625-1629.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 142]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
68.  Armuzzi A, Cremonini F, Bartolozzi F, Canducci F, Candelli M, Ojetti V, Cammarota G, Anti M, De Lorenzo A, Pola P. The effect of oral administration of Lactobacillus GG on antibiotic-associated gastrointestinal side-effects during Helicobacter pylori eradication therapy. Aliment Pharmacol Ther. 2001;15:163-169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 182]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
69.  Sýkora J, Valecková K, Amlerová J, Siala K, Dedek P, Watkins S, Varvarovská J, Stozický F, Pazdiora P, Schwarz J. Effects of a specially designed fermented milk product containing probiotic Lactobacillus casei DN-114 001 and the eradication of H. pylori in children: a prospective randomized double-blind study. J Clin Gastroenterol. 2005;39:692-698.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Szajewska H, Albrecht P, Topczewska-Cabanek A. Randomized, double-blind, placebo-controlled trial: effect of lactobacillus GG supplementation on Helicobacter pylori eradication rates and side effects during treatment in children. J Pediatr Gastroenterol Nutr. 2009;48:431-436.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 78]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
71.  Cremonini F, Di Caro S, Covino M, Armuzzi A, Gabrielli M, Santarelli L, Nista EC, Cammarota G, Gasbarrini G, Gasbarrini A. Effect of different probiotic preparations on anti-helicobacter pylori therapy-related side effects: a parallel group, triple blind, placebo-controlled study. Am J Gastroenterol. 2002;97:2744-2749.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.0]  [Reference Citation Analysis (0)]
72.  Ojetti V, Bruno G, Ainora ME, Gigante G, Rizzo G, Roccarina D, Gasbarrini A. Impact of Lactobacillus reuteri Supplementation on Anti-Helicobacter pylori Levofloxacin-Based Second-Line Therapy. Gastroenterol Res Pract. 2012;2012:740381.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Lionetti E, Miniello VL, Castellaneta SP, Magistá AM, de Canio A, Maurogiovanni G, Ierardi E, Cavallo L, Francavilla R. Lactobacillus reuteri therapy to reduce side-effects during anti-Helicobacter pylori treatment in children: a randomized placebo controlled trial. Aliment Pharmacol Ther. 2006;24:1461-1468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 115]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
74.  Emara MH, Mohamed SY, Abdel-Aziz HR. Lactobacillus reuteri in management of Helicobacter pylori infection in dyspeptic patients: a double-blind placebo-controlled randomized clinical trial. Therap Adv Gastroenterol. 2014;7:4-13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 50]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
75.  Sheu BS, Wu JJ, Lo CY, Wu HW, Chen JH, Lin YS, Lin MD. Impact of supplement with Lactobacillus- and Bifidobacterium-containing yogurt on triple therapy for Helicobacter pylori eradication. Aliment Pharmacol Ther. 2002;16:1669-1675.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 142]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
76.  Goldman CG, Barrado DA, Balcarce N, Rua EC, Oshiro M, Calcagno ML, Janjetic M, Fuda J, Weill R, Salgueiro MJ. Effect of a probiotic food as an adjuvant to triple therapy for eradication of Helicobacter pylori infection in children. Nutrition. 2006;22:984-988.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 63]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
77.  Deguchi R, Nakaminami H, Rimbara E, Noguchi N, Sasatsu M, Suzuki T, Matsushima M, Koike J, Igarashi M, Ozawa H. Effect of pretreatment with Lactobacillus gasseri OLL2716 on first-line Helicobacter pylori eradication therapy. J Gastroenterol Hepatol. 2012;27:888-892.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 51]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
78.  Nista EC, Candelli M, Cremonini F, Cazzato IA, Zocco MA, Franceschi F, Cammarota G, Gasbarrini G, Gasbarrini A. Bacillus clausii therapy to reduce side-effects of anti-Helicobacter pylori treatment: randomized, double-blind, placebo controlled trial. Aliment Pharmacol Ther. 2004;20:1181-1188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 119]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
79.  Cindoruk M, Erkan G, Karakan T, Dursun A, Unal S. Efficacy and safety of Saccharomyces boulardii in the 14-day triple anti-Helicobacter pylori therapy: a prospective randomized placebo-controlled double-blind study. Helicobacter. 2007;12:309-316.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 97]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
80.  Duman DG, Bor S, Ozütemiz O, Sahin T, Oğuz D, Iştan F, Vural T, Sandkci M, Işksal F, Simşek I. Efficacy and safety of Saccharomyces boulardii in prevention of antibiotic-associated diarrhoea due to Helicobacterpylori eradication. Eur J Gastroenterol Hepatol. 2005;17:1357-1361.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Hurduc V, Plesca D, Dragomir D, Sajin M, Vandenplas Y. A randomized, open trial evaluating the effect of Saccharomyces boulardii on the eradication rate of Helicobacter pylori infection in children. Acta Paediatr. 2009;98:127-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 59]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]