Join Us | Latest Articles | Contact

Journal Home


Editorial Board


Archive


Submit to this journal


Current issue

Journal of Clinical Gastroenterology and Treatment





DOI: 10.23937/2469-584X/1510011



Eradication of Helicobacter Pylori Infection: Past, Present and Future

Baryshnikova NV1*, Uspenskiy YP1 and Suvorov AN2


1Department of internal diseases, Pavlov First St-Petersburg State Medical University, Russian Federation
2Department of molocular microbiology, Science research Institute of Experimental Medicine, Russian Federation


*Corresponding author: Natalia Baryshnikova, MD, PhD, Department of internal diseases, Pavlov First St-Petersburg State Medical University, 14, Vavilovykh Str, 195257 St-Petersburg, Russian Federation, Tel: +7-921-301-33-77, E-mail: baryshnikova_nv@mail.ru
J Clin Gastroenterol Treat, JCGT-2-011, (Volume 2, Issue 1), Review Article; ISSN: 2469-584X
Received: November 18, 2015 | Accepted: January 23, 2016 | Published: January 26, 2016
Citation: Baryshnikova NV, Uspenskiy YP, Suvorov AN (2016) Eradication of Helicobacter Pylori Infection: Past, Present and Future. J Clin Gastroenterol Treat 2:011. 10.23937/2469-584X/1510011
Copyright: © 2016 Baryshnikova NV, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.



Abstract

This review is a comprehensive summary of different variants of anti-Helicobacter pylori therapy from past strategies to the current state of the art. Nowadays we see a progressive decreasing of eradication rate in many countries in case of use standard triple therapy. It can be associated with high clarithromycin resistance of Helicobacter pylori. Gradual increase in number of the used antibiotics, the increase in duration of treatment, use of new antibacterial compounds and schemes of treatment do not lead to a long-term positive effect on eradication rate and on preservation of risk of development of side reactions. It is necessary to pay active attention to new approaches to treatment and alternative options of therapy of an Helicobacter pylori infection. One of the most perspective methods of improving the efficacy of eradication can be the usage of probiotics, especially in addition to standard therapy. Probiotics have some mechanisms to influence on Helicobacter pylori: lactic acid production, synthesis of bacteriocins and antimicrobial metabolites, concurrence for adhesion sites, the reparation of the barrier function of the stomach mucosa, a decrease of inflammation and increase of immunity of infected humans. Bismuth subcitrate is very effective in eradication, cytoprotection, and atrophic changes regression and can be recommended for eradication schemes as classic quadrotherapy also as a 4th additional component in classic triple therapy.


Keywords

Helicobacter pylori, Eradication, Antibiotic resistance, Probiotics, Bismuth


Introduction

For last three decades, researchers consider Helicobacter pylori to be the hot topic and keep a strong focus on it. There are numerous lines of anti-H. pylori therapy. Since 80s up until now eradication therapy was constantly changing. It is being altered nowadays as well. Several lines of therapy didn’t establish appropriate efficacy when others remain to be highly effective up until now. Along with old methods, there are active discussions on implementing new compounds and alternative methods to impair on this infection.

The stages of development of eradication therapy could be divided into two sub-periods: before Maastricht and after Maastricht, between which Fourth Maastricht Consensus era is a real state of the art. We see the high probability for new methods of treatment to take a turn, many of which are under development nowadays.


Past Treatment Strategies

Before maastricht consensus period

B.J. Marshall and J.R. Warren were the first who could isolate a microorganism in human stomach mucosa and the first who could cultivate it in 1982 [1]. After that, these scientists made several additional experiments confirm an important role of Helicobacter pylori (Campylobacter pyloridis) in the pathogenesis of chronic gastritis and ulcer disease. The main experiment was performed by B.J. Marshall who drank liquid with pure culture of this microorganism and after 10 days he have a symptoms, endoscopic and morphological evidence of acute gastritis and H. pylori (C. pyloridis) in stomach mucosa [http://www.ncbi.nlm.nih.gov/pubmed/25167944]. After that B.J. Marshall eradicated H. pylori (C. pyloridis)-associated gastritis using bismuth and metronidazole [3]. J.R. Warren и B.J. Marshall could show that antibiotics are effective in majority of cases of chronic gastritis and duodenal ulcer [3,4].

So in was an onset of an active investigation of the efficacy of different antimicrobial drugs in H. pylori eradication with detection of antibiotic resistance of microorganism (Table 1).



Table 1: Primary H. pylori resistance to antibiotics [5]. View Table 1


Table 1. Primary H. pylori resistance to antibiotics [5].

The main cause for formation of the secondary resistance to antibiotics is a genetic factor: e.g. change of level of an expression of own genes due to the high genetic variability of H. pylori. Genetic changes of microbe can be associated with uncontrolled usage of antibiotics due to other diseases (e.g. usage of macrolides in patients with urogenital infections). Resistance to antibiotics is the important factor of low eradication rate. Already in B.J. Marshall work [4] it was revealed that all samples of a microorganism were sensitive to penicillin, erythromycin, tetracycline, cephalosporins, gentamicin and bismuth citrate and only 80% of the studied samples were sensitive to metronidazole or tinidazole [4]. At the time practitioners and researchers saw ways to overcome resistance to antibiotics by means of bismuth addition in schemes of treatment [6], which is actual method also nowadays.

Eradication therapy started as a monotherapy or double-antibiotic therapy.

Bismuth salts, especially colloid bismuth subcitrate, one of the first drugs which began to be used in H. pylori eradication [7,8]. Bismuth subsalicylate also have certain efficacy against this microbe: results of placebo-controlled trial shown that this drug suppressed growth of H. pylori in of 65% of patients and eradicated it in one patient [9].

The schemes which are containing bismuth and nitroimidazoles have shown significant clinical success. So, when comparing efficacy of different variants of an anti-Helicobacter therapy, it was shown that efficacy of H. pylori (C pyloridis) eradication scheme with colloidal bismuth subcitrate and tinidazole is more that 70% [10]. Schemes with omeprazole and bismuth also were in use at that time [11].

One of the first schemes of triple therapy included colloidal bismuth subcitrate, tetracycline and metronidazole with application period of 2-4 weeks. It was effective in 91-96% of patients [12,13]. Efficacy of triple therapy with bismuth, amoxicillin and metronidazole was higher than 80% [14].

Standard triple therapy: proton pump inhibitor (PPI), amoxicillin, and clarithromycin began to use in the middle of 90th in XX century [15]. Due to high efficacy this scheme was claimed as a gold standard for the treatment of H. pylori and became a worldwide recommended for first-line eradication [16]. In studies conducted during that time, standard triple therapy (7-14 days) was shown > 80% and even > 90% eradication success [17,18]. However, the subsequent efficiency of this treatment scheme decreased progressively. The core reason for such decrease is growth of H. pylori resistance to clarithromycin [16,19]. This research reflected the need for controlling level of H. pylori resistance to clarithromycin. It was recommended to avoid usage of this antibiotic if regional resistance level higher than 15-20% [20,21].

In the course of evolution of eradication therapy H2-blockers were replaced with the PPI which are possessing higher anti-acid activity and not having so expressed withdrawal syndrome that allowed them to have a primary position in different anti-H. pylori schemes [22].


Maastricht consensus period

Since today, there were four Maastricht Consensus: postulates of First Maastricht Consensus have been published in 1997, Second - in 2000, Third - in 2007 and Fourth - in 2012.

According to First Maastricht Consensus [17] for the therapy of H. pylori infection the triple therapy is recommended, consisting of a PPI, and two of the following: clarithromycin, a nitroimidazole (metronidazole or tinidazole) and amoxicillin. In various combinations it scheme was recommended to use as a first-line therapy. At that moment, triple therapy (bismuth tripotassium dicitrate plus metronidazole and tetracycline) have lower efficacy than PPI, clarithromycin, and amoxicillin therapy. It can be explained by two important mechanisms of PPI-antibiotic combination: anti-acid and antibacterial effects. It was recommended to use a standard dose of PPI (e.g. omeprazole 20 mg, lansoprazole 30 mg, pantoprazole 40 mg [23]), twice a day and antibiotic combinations during 7 days such as:
a. metronidazole 400 mg twice daily (or tinidazole 500 mg twice daily) plus clarithromycin 250 mg twice daily;
b. amoxicillin 1000 mg twice daily plus clarithromycin 500 mg twice daily (advisable when metronidazole resistance is likely)
c. amoxicillin 500 mg three times daily plus metronidazole 400 mg three times daily (advisable when clarithromycin resistance is likely).

In case of no efficacy of first-line therapy, a second-line therapy should be selected after consideration of previous treatment and microbial sensitivities.

According to Second Maastricht Consensus [20] for the first-line therapy of H. pylori infection it was recommended triple therapy using a PPI or ranitidine bismuth citrate in standard dose (e.g. esomeprazole 20 mg, lansoprazole 30 mg, omeprazole 20 mg, pantoprazole 40 mg, rabeprazole 20 mg, ranitidine bismuth citrate 400 mg) with clarithromycin (500 mg twice a day) and amoxicillin (1000 mg twice a day) or metronidazole (500 mg twice a day) for 7 days or longer (10-14 days). An H2-blocker ranitidine bismuth citrate was included in eradication scheme because numerous studies demonstrating similar efficacy of ranitidine and PPIs [24,25]. Second-line therapy should use quadruple therapy with a PPI in standard dose, bismuth (120 mg four times a day), metronidazole (500 mg twice a day) and tetracycline (500 mg four times a day). Where bismuth is not available, second-line therapy should be performed by PPI triple therapy [20].

It is interesting that already in this period spoke about possibilities of probiotics and vaccines against H. pylori infection. Also at that time it was postulated about the importance of H. pylori resistance to clarithromycin as a factor of low eradication rate [26]. In case of high metronidazole resistance, it was recommended to use furazolidone [27].

According to Third Maastricht Consensus [21] for the therapy of H. pylori infection it was still recommended PPI-clarithromycin - amoxicillin - or metronidazole regimen, but only if the primary resistance to clarithromycin in the area is lower than 15% to 20% and prevalence of metronidazole resistance is lower than 40%. Several researchers have revealed that a 14-day rather than a seven-day treatment had a slight advantage in terms of treatment success [21]. Bismuth-based quadruple therapies (when available) are acceptable as alternative first-line therapy. For second-line therapy, bismuth-based quadruple therapies or PPI-amoxicillin or tetracycline and metronidazole are recommended. Third line therapy or rescue therapy is used after the determination of sensitivity of H. pylori to antibiotics.


Present treatment strategies

According to Fourth Maastricht Consensus [28] the following variants of eradication therapy showed the efficiency in large-scale, placebo-controlled researches (duration 7-14 days) and are recommended for clinical application:
1. The triple therapy including PPI plus clarithromycin and amoxicillin or metronidazole are still in use. The most recent data shows this combination has lost some efficacy and often allows the cure less than the 80% of patients [29].
2. Sequential treatment includes a 5-day period with PPI and amoxicillin, followed by a 5-day period with PPI plus clarithromycin and metronidazole (or tinidazole) in standard doses [30].
3. Three antibiotics (e.g. amoxicillin, calrithromycin and metronidazole) together with a PPI (non-bismuth quadruple therapy or concomitant therapy) [31,32].
4. The bismuth-containing quadruple therapy including PPI , bismuth salts, tetracycline and metronidazole esspessially in the same pill [33].
5. It is possible to use the triple therapy on the base of a levofloxacin (IPP, amoxicillin, levofloxacin) as an alternative scheme of treatment of rescue therapy [28].

Modern approach also suggests hybrid therapy: a 5-7-days dual therapy with a PPI (standard dose, twice a day) and amoxicillin (1000 mg, twice a day) followed by a 5-7-days quadruple therapy with a PPI (standard dose, twice a day), amoxicillin (1000 mg, twice a day), clarithromycin (500 mg, twice a day) and metronidazole (500 mg, twice a day) [34].

As one can see over the time the number of antibiotics and duration of treatment are progressively increasing. This can be a reason of raise in frequency of side effects of eradication such as colon dysbiosis and decrease of eradication rate due to formation of antibiotic resistance.

There are several problems that can reduce the efficacy of anti-H. pylori therapy (e.g. resistance to antibiotics, low patients compliance, usage of generics, etc.). One of the main factors of lower eradication success is a progressive growth of microorganism resistance to antibiotic especially to clarithromycin. It is revealed that high resistance of H. pylori to clarithromycin leads to catastrophic decrease of eradication therapy efficacy from 80-90% to 30-60% [35]. Summarized results of 20 European studies about efficacy of first-line therapy (PPI, amoxicillin and clarithromycin) in 2751 patients show that in case of clarithromycin resistance eradication rate decrease to 18,3% (in compare with 87,8% in case of infect of non-resistance H. pylori strains [36]. Nowadays we see worldwide tendency to growth H. pylori clarithromycin resistance (Table 2).



Table 2: The level of H. pylori resistance to clarithromycin (According recent Workshop of European Helicobacter Study) [37]. View Table 2


Table 2. The level of H. pylori resistance to clarithromycin (According recent Workshop of European Helicobacter Study) [37].

Significantly different results of H. pylori clarithromycin resistance are a way to differ treatment in various regions. For optimization of eradication schemes, Fourth Maastricht Consensus recommended different therapy strategies in regions with high and low clarithromycin resistance [28].

Progressive decreasing of standard triple therapy efficacy in many countries all around the world is a reason to find possible ways to improve the efficacy of anti-H. pylori therapy. These ways can be [28,53]:
1. Usage of high dosage of PPI
2. Prolonged eradication therapy
3. High dosage of traditional used antibiotics
4. Change of antibiotics with known high H. pylori resistance to new antibacterial drug or drugs with low H. pylori resistance.
5. Sequential treatment, concomitant and hybrid therapy
6. Additional usage of probiotics


High dosage of proton pump inhibitors application

Increasing the dosage of PPI causes concentration rise of active component in the blood stream, as well as an antisecretory effect. It can increase an eradication rate of 6-10% [54]. Serious side effects while the high dosage of PPI for eradication therapy does not get developed. The reason is a very limited time of high dosage application. However, increased dosage of antisecretory therapeutics is not commonly approved. The reasons are non-optimal price/quality ratio and lack of effect on complications decrease, i.e. bleeding/hemorrhage [55].


Prolonged of eradication therapy

This is the most widely spread and best-adopted option of increasing efficacy of eradication therapy. As a part of Standards, the method provides legal reliability for practical physicians for cases of fail and patient objections, complications manifestations, and other situations. Currently, according to Fourth Maastricht Consensus, an extension of anti-H. pylori therapy up to 10-14 days increases the efficacy of eradication by 5% (in comparison to 7 days treatment) [28]. The study by a research group in Italy provided evidence on the higher efficacy of 14 day combined therapy (proton pump inhibitors, amoxicillin, clarithromycin) compared to 7-day treatment length (70% and 57% accordingly) [56]. Research group from Croatia provided a comparison of 7-, 10- and 14-day treatment schemes. Results indicated proven efficacy over 80% for combination of proton pump inhibitors, amoxicillin, clarithromycin only for 10 and 14 days length. In case of combination proton pump inhibitors, amoxicillin and metronidazole efficacy of 80% was reached only with 14-day length of therapy [57].

The meta-analysis comparison for 2-week and 1-week eradication therapy research for non-Russia practices confirms that the extension of therapy length allows to reach better eradication results [58]. Russian studies also confirm the necessity of length extension of anti-Helicobacter therapy because according to the modern state of the art seven-day three-component scheme is not effective enough [59].


Increasing the dosage of traditional antibacterial medicaments

The dynamics of dosage increasing for eradication therapy of H. pylori could be observed through the development of Maastricht protocols/consensus. Through the period between First and Third Maastricht consensus the doses of Metronidazole were increased from 400 mg 2 times a day up to 500 mg 3 times a day, amoxicillin from 500 mg 3 times a day to 1000 mg 2 times a day, clarithromycin from 250 mg 2 times a day to 500 mg 2 times a day [21].

Unfortunately, this way of increasing of eradication rate can lead to developing of complications, in particular, a colon dysbiosis but doesn't allow increasing the percent of a successful eradication significantly.


Usage of new antibacterial medicaments

Levofloxacin-based therapy is an effective in H. pylori eradication [60]. In the majority of studies use a scheme: PPI in standard dose, amoxicillin 1000 mg, levofloxacin 500 mg twice a day 10-14 days as a first-line, second-line or third-line therapy. Unfortunately in some countries there is data indicating that usage of levofloxacin leads to the progressive growth of H. pylori resistance to fluoroquinolones. It is limited of levofloxacin-based therapy. There are data revealing efficacy of other fluoroquinolones in eradication schemes, e.g. moxifloxacin [61] sitafloxacin [62].

Some macrolides other than clarithromycine demonstrate high efficacy in eradication schemes, e.g. azithromycin and josamycin. In some studies it was shown that scheme with azithromycin has eradication rate around 78% - 86.3%, with josamycin - в 85.6%, with josamycin and bismuth - more than 90% [63].

Nowadays some scientists use anti -TB-antibiotic rifabutin [60] in the scheme: PPI, amoxicillin 1000 mg, rifabutin 150 mg twice a day 10-14 days. Efficacy of this treatment is around 79%-95% as a second-line therapy and 61%-68% as a third-line therapy [60]. However, often side reactions of rifabutin, e.g. myelotoxicity, thrombocytopenia, disorders of vision, can limited usage of this scheme [60]. Moreover wide use of rifabutin can lead to increase the percent of rifabutin-resistant strains Mycobacterium tuberculosis, which significantly worsen treatment of patients with tuberculosis.

Nitrofurans are also effective in eradication schemes because H. pylori resistance to this group of antimicrobial medicaments is low. Usually they use in case of methronidazole-resistant (nitromidazole-resistant) strains of H. pylori. Furazolidone-based schemes were offer for recommendation in China gastroenterological society due to its high efficacy in China [64]. However, usage of furazolidone has some restrictions. Important of these restrictions are side effects, e.g. gepatotoxity, neurotoxity, gematotoxity that limited an administration of furazolidone. In some works scientists use eradication scheme with another nitrofuran - nifuroxazide. Efficacy of these schemes is around [53]. Nowadays an optimal nitrofuran for treatment of H. pylori-associated diseases is nufuratel. Efficacy of nifuratel-contain eradication schemes is around 82-100% [65]. Additional positive feature of nifuratel is correction of colon microflora with decrease content of semi-pathogenic microorganism and increase of Bifidobacteria spp. and Lactobacillus spp. level [65].

Rifaximin also used in the second line of eradication therapy in dose 400 mg 2-3 times a day. Scientists from Italy used PPI, clarithromycin and rifaximin in treatment of 24 patients and see eradication rate 58% [66].

Some groups attempted to use doxycycline in schemes of an eradiсation therapy [67,68].


Usage of bismuth salt

Due to the increasing resistance of H. pylori to сlarithromycin and other antibiotics in eradication schemes include salt of bismuth because of absence of primary and secondary resistance of H. pylori to bismuth. Also usage of bismuth can protect patients against side effects of treatment such as antibiotic-associated diarrhea and colon dysbiosis. Moreover, there are data that bismuth tripotassium dicitrate have a beneficial effect on a content of an intestinal microflora (as an intestinal antiseptic) [69]. In some cases (e.g. at elderly people of allergic patients) it is possible to use a double therapy (PPI and bismuth threepotassium dicitrate) or monotherapy with bismuth [70].

In a number of research works it scientists obtained data that addition of a bismuth tripotassium dicitrate to triple standard therapy promotes reliable increase in percent of an effective eradication [71-73]. In this case monitoring of the growing resistance to a clarithromycin isn't required and bismuth-containing therapy can compensate the lack of new, alternative antibiotics.

Cytoprotective properties of bismuth provide effective protection for stomach mucosa against the damaging action of products of an inflammation for the purpose of prevention of progressing of gastritis [74]. It is shown, that usage of bismuth salt (e.g. tripotassium dicitratobismuthate) leads to regression of atrophic changes of a stomach [75].


Sequential therapy

Sequential therapy is an alternative for classic triple standard therapy [76]. The main goal of this variant of treatment is overcoming of the increasing resistance of H. pylori to a clarithromycin. Therefore, this concept in the treatment of an H. pylori infection can be recognized by the new standard, especially in the countries and regions with the high resistance of a microorganism to macrolides. Sequential therapy includes two parts: 1st part - usage of PPI and amoxicillin in standard doses for five days, 2nd part - usage of PPI, clarithromycin and metronidazole (or tinidazole) also for five days. It is possible to add salt of bismuth to this therapy. All mechanisms of high efficiency of sequential therapy aren't clear yet. Possibly, usage of amoxicillin leads to 1. "weakening" of a cellular wall of bacteria that interferes with formation of the channels blocking action of a clarithromycin and causing resistance of a bacterium to it and 2. promotes development of most expressed pharmacological effect of the antibiotics accepted in the 2nd phase. Perhaps one of the reasons of higher efficiency of sequential therapy, is combination of medication with an additional antibacterial drugs (tinidazole or metronidazole) [76]. According to results of some studies, efficiency of sequential therapy doesn't depend on properties of a microbe (cagA+status, bacterial loading) and the human (associated diseases, smoking, etc.) [77].


Concomitant therapy

Concomitant therapy includes four compounds: 20-40 mg of PPI twice daily and 1000 mg of amoxicillin twice daily, 500 mg of clarithromycin twice daily 500 mg of metronidazole every eight hours for 10-14 days. This scheme of therapy is recommended in case of high clarithromycin resistance. The efficacy of this treatment method is confirmed by results of meta-analysis. Seven studies provided data on 2412 adult patients shown that there are no significant differences between concomitant therapy and sequential therapy. Also, there was no difference in the rate of adverse events [78]. Other meta-analysis and clinical trials show that concomitant therapy is more effective that sequential or hydrid therapy, e.g. 10-day concomitant therapy showed better eradication rate than sequential therapy (94.4% v.s. 82.2%, p = 0.002) [79]. In other study was revealed that triple therapy eradication rates were 76.2%-84.2%; 84.4% - in sequential therapy and 94.4% in the concomitant group (P = 0.0002) [80].


Hybrid therapy

Hybrid therapy is the therapy consisting of PPI in standard dose, amoxicillin 1000 mg both twice daily for 10-14 days; plus clarithromycin, 500 mg and tinidazole 500 mg both twice daily just during the last 5-7 days [81,82]. This type of eradication scheme has lower efficacy in compare with sequential and concomitant therapy. But some studies showed similar efficacy [83,84].


Usage of probiotics

One of the most perspective ways for optimization and improving eradication therapy is the usage of probiotics in addition to standard schemes. Several years ago it was only an interesting idea but nowadays it often use and has promising results in the majority of studies. In Fourth Maastricht Consensus, it is postulated that certain probiotics and prebiotics show promising results as an adjuvant treatment in reducing side effects [28].

Probiotics have some mechanisms to influence on H. pylori: lactic acid production, synthesis of bacteriocins and antimicrobial metabolites [85]. Other possible mechanisms of probiotics are concurrence for adhesion sites, the reparation of the barrier function of the stomach mucosa, a decrease of inflammation and increase of immunity of infected humans [53,86].

There are promising results for evidence that probiotics decrease of H. pylori colonization of the gastric mucosa. Results of meta-analysis showed that usage of probiotics in addition to standard eradication therapy lead to an increase of eradication rate and decrease of side effects frequency (Table 3) [87-89].



Table 3: A meta-analysises that assessed the impact of different probiotics in addition to H. pylori eradication therapy [87]. View Table 3


In some studies is shown that standard triple therapy with bismuth and probiotics is very effective. For example, usage of 30 mg lansoprazole twice daily, 1 g amoxicillin twice daily, 1 g clarithromycin once daily and 1,048 mg bismuth subsalicylate twice daily for 7 or 14 days and probiotic bacteria composed of Bifidobacterium lactis, Lactobacillus acidophilus and Lactobacillus paracasei have 100% efficacy against H. pylori [97].

In work of Turkey scientists was no statistically significant efficacy of probiotics against H. pylori in comparison with placebo [98].

A meta-analysis from China shows that usage of probiotics for H. pylori eradication can increase eradication rates and reduce side effects of antibiotics. Probiotic administration prior and subsequent to the therapy and for a duration of > 2 weeks may increase the eradication efficacy. According to this meta-analysis, the most effective probiotics are Lactobacillus or multiple probiotic strains [99].

In Russian practice there is also use of different variants of probiotics therapy: pre-eradication therapy - 3-4 weeks before eradication to improve immunity of patients and increase of predictability of an eradication success; co-eradication therapy - at the same time with eradication to increase of eradication rate and decrease of side effects frequency; post-eradication therapy - 3-4 weeks after eradication for intestinal microflora correction and decrease of probability of H. pylori reinfection (recolonization) [53].


Probiotic monotherapy

This method of treatment of H. pylori infection can be recommended for some categories of patients: patients with non-atrophic chronic gastritis and/or duodenal ulcer in remission; patients infected with low virulent strains of H. pylori; patients who have allergic reaction to antibiotics; patients who refuse to take antibiotics; persons who infected with H. pylori but haven’t any clinical symptoms of dyspepsia; for family members of H. pylori infected patient for prophylaxis against microbe invasion. There are many studies confirming the efficacy of probiotics monotherapy for H. pylori eradication. This therapy demonstrates lower efficacy that standard schemes but it have a very low frequency of side effects. Also in the era of increasing of H. pylori antibiotic resistance probiotic therapy can be a good alternative to classic treatment. For better effect, the duration of probiotic therapy can be 3-4 weeks or longer.

Usage of probiotic which contain lacto-acid bacteria in the treatment of chronic H. pylori associated gastritis patients eradication rate was 48% in one study [100] and 41% in another study [101]. Eradication was successful in 6 from 14 patients who used L. acidophilus [102]. In other study it was shown that 29 patients with H. pylori associated ulcer disease who used Enterococcus faecium strain L-3 106-7 cfu/g 3 dragee 3 times a day during one month have success of eradication 38% [103]. These results were significantly higher than level of spontaneous H. pylori eradication (approximately 5%) and can be explained by improving of colon microbiota balance, positive influence of probiotic on the human immune system or by the direct inhibition of H. pylori by bacteriocins of probiotic strain. In this study in case of 7-days standard triple therapy (control group) eradication rate was 60% [103].

There are several pieces of evidence of the role of probiotics against the carcinogens of H. pylori. It is possible that probiotics may act as antineoplastic agents in the stomach by affecting the polyamine content and functions [104].

The results of in vitro studies demonstrated that probiotics can directly inhibit H. pylori [105,106]. This is very important for future probiotics usage as anti-H. pylori agents. It was shown that 14 strains of Helicobacter pylori were successfully cultivated from dyspeptic patients. Incubation was made in standard conditions for H. pylori. Two variants of probiotic medications were used: 1st contain Enterococcus faecium strain L-3, 2nd - lyophilisate cultural fluid of Bacillus subtilis. The studied probiotic medications were dissolved in distilled water in part 1:100 and were added in a cup with an agar with different H. pylori strains. The assessment of growth of H. pylori was analyzed after 6-7 days. Inhibition of grow of H. pylori was in 50% cases with Bacillus subtilis and in 78.6% with Enterococcus faecium strain L-3 [106].

Despite the rather large number of the research works confirming the efficiency of probiotics in eradication, larger randomized controlled investigations are needed to understand clearly the effects of probiotics on H. pylori eradication.


Future Treatment Strategies

Metabiotics

Recent researches shown innovative and a very actual approach is the usage of probiotic metabiotics - medicaments on base of products of a metabolism or structural components of probiotic microorganisms [107].

Very promising application as a way of elimination of H. pylori from a human organism is use of a medicament on a base of the inactivated cells of pro-biotic bacteria of Lactobacillus reuteri DSMZ 17648 (Pylopass™) allocated and processed in the biotechnological way. Lactobacillus reuteri DSMZ 17648 - special strain of lactobacilli possessing unique ability: specific contact cells of H. pylori and to form co-aggregans without influencing other bacteria and normal intestinal flora. This specific co-aggregation decrease of H. pylori mobility. Also co-aggregants of microbe aggregants of pathogens cease to communicate with mucous a digestive tract and "are washed away" from a stomach that as a result leads to reduction of colonization of H. pylori in a stomach mucosa, reducing risk of development of gastritis and ulcer disease [108,109].


Bacteriophages

Bacteriophages are in a focus of interest of a great number of scientists: they in compare with probiotic microorganisms possess the highest degree of specificity concerning certain microorganisms. But also it is a negative side: high variability of microorganism leads to loss of specificity of bacteriophages, and, therefore, and their "uselessness" in this case. In addition there are difficulties of selection and reproduction of phages, and the immune system can react to this type of treatment also negatively.


Bacteriocins

Bacteriocins (bacterial proteins or peptides with antimicrobial action) is one of the most interesting for investigation due to presence in bacteriocins specify of bacteriophages and safety of probiotics [110]. Nowadays bacteriophages are the factors of microbial antagonism. They provide the regulation of bacterial population a colonization resistance of humans and animals to pathogens. Almost all bacteria from lactic acid bacteria can made bacteriocins. For example, In Korean study antimicrobial activity of seven bacteriocins (nisin A; lacticin A164, BH5, JW3, and NK24; pediocin PO2; and leucocin K) produced by lactic acid bacteria against H. pylori strains was investigated in vitro using a broth microdilution assay. Lacticins A164 and BH5 showed the strongest antibacterial activity against H. pylori strains [111].


Vaccines

Another method to fight with this microorganism is attempted to create a vaccine against H. pylori. Allegedly, process of increasing of content of immunocompetent cells in a stomach mucosa at early stages of pathogenesis of H. pylori-associated diseases leads to exhaustion of mechanisms of protection and dictates need to stimulate development of protection factors by immunization [112]. After vaccination, a specific immune answer develops due to the synthesis of antibodies, and the combined cellular, molecular and humoral answer also develop that have to provide full protection against H. pylori. The most effective ways of vaccination are intramuscular and intranasal, but intragastric and rectal are less effective [113].

As antigens for immunization against H. pylori can be used several factors of pathogenesis of H pylori-associated diseases: VacA, CagA, NapA, BabA, SabA and urease [114]. The majority of research works is conducted on animals, mainly on mice, however, optimum strategy of creation of vaccines against a H. pylori infection is strategy of possibility of use of the developed vaccines at people [115]. Promising research is the study of Malfertheiner P. in which studied safety and an immunogenetics of the vaccine for intramuscular vaccination with recombinant VacA, CagA and NapA on aluminum hydroxide at 57 healthy H. pylori-negative volunteers in randomized blind research of the first phase with various terms and dosages of a vaccine. All persons who were vaccinated have been examined in 5 and 36 months after vaccination, in 18 and 24 months after the first vaccination. As a result, it was established that in all variants of immunization there was a development of specific IgG and activation of the cellular answer concerning one or two proteins and in 86% of cases - all three anti-genes. Thus, the number of side effects was little [116].

Xu C et al. created a recombinant strain of Salmonella, which express H. pylori urease and interleucin-2. At immunized mice were a more significant decrease of results of rapid urease test in comparison with mice, which were immunized with the vaccine, expressing only urease [117]. The level of interferon-γ and defensine-1 in stomach increase after immunization. It can be an protect factor against H. pylori aggression [112].

Also promising results show the usage by upon oral immunization of recombinant Bacillus subtilis sporeswith express of UreB protein of Helicobacter acinonychis [118]. In another study is revealed that the recombinant L. lactic expressing UreB can be potentially used as an edible vaccine for controlling H. pylori infection [119,120].

Efficacy of DNA-vaccines also confirmed in mice (vaccination on base of urease B) [121]. Potential advantages of DNA vaccines are simplicity of preparation and use (can be used together with food), temperature stability and stimulation of the cytotoxic T-cellular answer that is important for treatment and prevention of a H. pylori-associated diseases [122]. Mechanisms of activity of DNA vaccines are studied insufficiently. Theoretically, they are safe, but it isn't necessary to forget that the integration of DNA into cages of a macroorganism can promote development of anti-DNA - autoimmune reaction.

Unfortunately, the vaccine, which could be recommended for use at the person, doesn't exist yet, despite more than 20-year history of their creation and a large number of examples of efficiency of vaccines at animals. Mechanisms of action of vaccines at animals and the person are clear insufficiently and need further specification. It isn't necessary to forget that side effects of vaccination against H. pylori aren't completely studied that also demands improvement of methods of creation of vaccines.


New acid blockers

New acid blocker Vonoprazan - potassium-competitive acid blocker (P-CAB) comes in gastroenterology around one year ago [123]. Now this medicine is approved in Japan for the treatment of acid-related diseases and H. pylori eradication. Vonoprazan is very strong acid secretion inhibitor and shows promising results of H. pylori eradication with vonoprazan-based triple therapy after failure of proton pump inhibitor-based triple therapy [124].


Alternative methods of H. pylori eradication

The main alternative ways of H. pylori eradication are usage of new medicaments and biologically active substances, ozonetherapy, laserotherapy. Eradication rate in case of using propolis can be around 60% [125,126]. In is shown good efficacy by melatonin for increase of eradication rate [127]. Ozone seems to be effective in treatment f H. pylori-associated gastritis and ulcer disease due to bactericide properties, anti-inflammatory effect (oxidation of arachidonic acid - predecessor of prostaglandin E starting inflammatory process); immunomodulatory action, and also analgesic effect [128,129].

Treatment with use of the laser can increase the effect of antibacterial medicaments that leads to high efficacy of H. pylori eradication (86.7% in case of use antibiotics and laser in comparison with 66.7% in case of use antibiotic only) [130]. After exposure of helium-neonic laser contours of an external and internal membrane of H. pylori lost the clearness, and on separate sites were faltering or completely disappeared. It confirm the direct properties action of laser on this microorganism [131]. Using laserotherapy it is possible to decrease inflammatory and destruction (ulcerate) lesions and increase eradication rate. After using laserotherapy (endocscopic) for three times negative result of rapid urease test was 85.8% in antrum and 88.5% in stomach corpus. Histological data shown that after treatment with laserotherapy H. pylori absences in 71.4% in antrum and 77.4% in stomach corpus [132].


Role of genetic features of H. pylori

Genetic properties of H. pylori can play an important role in the efficacy of eradication. It is established that cagA-positive strains are more susceptibility to antibiotics in comparison with cagA-negative strains. Presence of allele VacA s1m1 also increases the susceptibility of microorganism in comparison with allele VacA s2m2 [133]. Only the minor part of H. pylori-infected people (less than 10-20%) has H. pylori-associated diseases [134]. This supervision is explained by the fact that population of H. pylori possesses high heterogeneity, and its’ strains considerably differ in a virulence. Based on mentioned clinical manifestations of diseases are capable to cause not all of H.pylori strains [135]. Since nineties of XX century it was known H. pylori strains were different on the genome and called "ulcerogenic" (synthesis cytotoxins, are associated with stomach ulcer, active or atrophic gastritis) and "non-ulcerogenic" (don't synthesis cytotoxins, are associated with simple gastritis) [136,137]. It will become possible to choose the type of treatment on the basis of such important factor as genetic features of strains of H. pylori. Thus, it is necessary to notice that in a case of infection of the patient with low virulent strains also possibly monotherapy use by a probiotics or metabiotics as an alternative to antibiotics and PPI.


Role of genetic properties of human

There are an additional factor that influence on eradication efficacy is human genetic features especially polymorphism of CYP2C19. The CYP2C19 genotype is an important factor of H. pylori eradication in patients taking omeprazole-based or lansoprazole-based triple therapies and levofloxacin-based triple therapy but no so significant in case of rabeprazole-based or esomeprazole-based triple therapies [138,139]. Mutations in CYP2C19 influence on metabolism of different drugs and divided on three types: without mutations - rapid metabolizers, a mutation in one allele - intermediate metabolizers, mutations in two alleles - slow metabolizers. Eradication rate is the highest in slow metabolizers [140]. The CYP2C19 and IL-1B-31CC genotype can be predictors of success of treatment [141]. Also polymorphism of IL-1β-511 significantly influence on eradication rate: in T-allele (IL-1β-511 С/T or IL-1β-511 T/T) presence eradication have better results than in case of IL-1β-511 С/C allele [142].

Expression and functional activity of polyspecific ATP-dependent efflux transporter - P-glycoprotein (P-gp) influence of adsorption of many medicaments also play a role in the pathogenesis of H. pylori infection [143]. This expression and functional activity encoding by MDR1 (ABCB1) gene [144]. It is considered that genotypes of MDR1 3435 С/T and С/С are characterized by the high and moderate level of an expression of P-gp on apical poles of membranes of intestinal enterocytes. In turn, the genotype of MDR1 3435 T/T is associated with the low level of an expression of P-gp that causes higher level of absorption of medicinal substance in a system blood-groove in comparison with genotypes of C/T and C/C [145]. Unfortunately the current usage of genetic analysis in H. pylori infected patients is not common in clinical practice. It can be explained by high cost of the analysis and absence of diagnostic standards.

In the conclusion, it is necessary to notice that today anti-H. pylori therapy with different antibiotics using at the same time cannot be dogmatic. It is important to investigate genetic features of microorganism and human. It is possible that increase of treatment duration, an increase of doses of antibiotics, usage of new antibiotics none enough effective. A large number of researches in the different countries of the world will help to develop optimum ways of an eradication of H. pylori. Considering that gradual increase in number of the used antibiotics, the increase in duration of treatment, use of new compounds and schemes of treatment do not lead to a long-term positive effect on preservation of risk of development of side reactions, it is necessary to pay active attention to new approaches to treatment and alternative options of therapy of an H. pylori infection.


References
  1. (1983) Unidentified curved bacilli on gastric epithelium in active chronic gastritis. Lancet 1: 1273-1275.

  2. Marshall BJ, Armstrong JA, McGechie DB, Glancy RJ (1985) Attempt to fulfil Koch's postulates for pyloric Campylobacter. J Aust 142: 436-439.

  3. Marshall BJ, Armstrong JA, Francis GJ, Nokes NT, Wee SH (1987) Antibacterial action of bismuth in relation to Campylobacter pyloridis colonization and gastritis. Digestion 37: 16-30.

  4. Marshall BJ, McGechie DB, Rogers PA, Glancy RJ (1985) Pyloric Campylobacter infection and gastroduodenal disease. J Aust 142: 439-444.

  5. Sidorenko SV (2015) Diagnostic and treatment of Helicobacter pylori-associated infections.

  6. Goodwin CS, Marshall BJ, Blincow ED, Wilson DH, Blackbourn S, et al. (1988) Prevention of nitroimidazole resistance in Campylobacter pylori by coadministration of colloidal bismuth subcitrate: clinical and in vitro studies. J Clin Pathol 41: 207-210.

  7. Goodwin CS, Marshall BJ, Blackbourn SJ, Warren JR, et al. (1989) Colloidal bismuth subcitrate (DE-NOL) and tinidazole healed duodenal ulceration with a low relapse rate due to elimination of Campylobacter pylori. J Chemother 4: 838-839.

  8. Miller JP (1989) Colloidal bismuth in the treatment of duodenal ulceration: the benefit for the patient. Scand J Gastroenterol Suppl 157: 16-20.

  9. Marshall BJ, Valenzuela JE, McCallum RW, Dooley CP, Guerrant RL, et al. (1993) Bismuth subsalicylate suppression of Helicobacter pylori in non-ulcer dyspepsia: a double-blind placebo-controlled trial. Dig Dis Sci 38: 1674-1680.

  10. Marshall BJ, Goodwin CS, Warren JR, Murray R, Blincow ED, et al. (1988) Prospective double-blind trial of duodenal ulcer relapse after eradication of Campylobacter pylori. Lancet 2: 1437-1442.

  11. Ciccaglione AF, Cellini L, Grossi L, Manzoli L, Marzio L (2015) A Triple and Quadruple Therapy with Doxycycline and Bismuth for First-Line Treatment of Helicobacter pylori Infection: A Pilot Study. Helicobacter 20: 390-396.

  12. George LL, Borody TJ, Andrews P, Devine M, Moore-Jones D, et al. (1990) Cure of duodenal ulcer after eradication of Helicobacter pylori. J Aust 153: 145-149.

  13. Iser JH, Buttigieg RJ, Iseli A (1994) Low dose, short duration therapy for the eradication of Helicobacter pylori in patients with duodenal ulcer. J Aust 160: 192-196.

  14. Wagner S, Varrentrapp M, Haruma K, Lange P, Müller MJ, et al. (1991) The role of omeprazole (40 mg) in the treatment of gastric Helicobacter pylori infection. Z Gastroenterol 29: 595-598.

  15. Bazzoli F, Pozzato P (1997) Therapy of H. pylori infection. J Physiol Pharmacol 48 Suppl 4: 39-46.

  16. Papastergiou V, Georgopoulos SD, Karatapanis S (2014) Treatment of Helicobacter pylori infection: Past, present and future. World J Gastrointest Pathophysiol 5: 392-399.

  17. (1997) Current European concepts in the management of Helicobacter pylori infection. The Maastricht Consensus Report. European Helicobacter Pylori Study Group. Gut 41: 8-13.

  18. Lind T, Veldhuyzen van Zanten S, Unge P, Spiller R, Bayerdörffer E, et al. (1996) Eradication of Helicobacter pylori using one-week triple therapies combining omeprazole with two antimicrobials: the MACH I Study. Helicobacter 1: 138-144.

  19. Fischbach L, Evans EL (2007) Meta-analysis: the effect of antibiotic resistance status on the efficacy of triple and quadruple first-line therapies for Helicobacter pylori. Aliment Pharmacol Ther 26: 343-357.

  20. Malfertheiner P, Mégraud F, O'Morain C, Hungin AP, Jones R, et al. (2002) Current concepts in the management of Helicobacter pylori infection--the Maastricht 2-2000 Consensus Report. Aliment Pharmacol Ther 16: 167-180.

  21. Malfertheiner P, Megraud F, O'Morain C, Bazzoli F, El-Omar E, et al. (2007) Current concepts in the management of Helicobacter pylori infection: the Maastricht III Consensus Report. Gut 56: 772-781.

  22. Lamouliatte H (1995) Adjuvant therapy for Helicobacter pylori eradication: role of lansoprazole in clinical studies. J Clin Gastroenterol 20 Suppl 1: S28-31.

  23. Labenz J, Tillenburg B, Weismuller J, Lutke A, Stolte M (1997) Efficacy and tolerability of a one-week triple therapy consisting of pantoprazole, clarithromycin and amoxycillin for cure of Helicobacter pylori infection in patients with duodenal ulcer. Aliment Pharmacol Ther 11: 95-100.

  24. Gisbert JP, Pajares JM, Valle J (1999) Ranitidine bismuth citrate therapy regimens for treatment of Helicobacter pylori infection: a review. Helicobacter 4: 58-66.

  25. Pipkin GA, Williamson R, Wood JR (1998) Review article: one-week clarithromycin triple therapy regimens for eradication of Helicobacter pylori. Aliment Pharmacol Ther 12: 823-837.

  26. Coelho LG, León-Barúa R, Quigley EM (2000) Latin-American Consensus Conference on Helicobacter pylori infection. Latin-American National Gastroenterological Societies affiliated with the Inter-American Association of Gastroenterology (AIGE). Am J Gastroenterol 95: 2688-2691.

  27. Mégraud F, Marshall BJ (2000) How to treat Helicobacter pylori. First-line, second-line, and future therapies. Gastroenterol Clin North Am 29: 759-773, vii.

  28. Malfertheiner P, Megraud F, O’Morain CA, Atherton J, Axon AT, et al. (2012) Management of Helicobacter pylori infection--the Maastricht IV/ Florence Consensus Report. Gut 61: 646-664.

  29. Graham DY, Fischbach L (2010) Helicobacter pylori treatment in the era of increasing antibiotic resistance. Gut 59: 1143-1153.

  30. Gisbert JP, Calvet X, O'Connor A, Mégraud F, O'Morain CA (2010) Sequential therapy for Helicobacter pylori eradication: a critical review. J Clin Gastroenterol 44: 313-325.

  31. Essa AS, Kramer JR, Graham DY, Treiber G (2009) Meta-analysis: four-drug, three-antibiotic, non-bismuth-containing “concomitant therapy” versus triple therapy for Helicobacter pylori eradication. Helicobacter 14: 109-118.

  32. Gisbert JP, Calvet X (2011) Review article: non-bismuth quadruple (concomitant) therapy for eradication of Helicobater pylori. Aliment Pharmacol Ther 34: 604-617.

  33. Malfertheiner P, Bazzoli F, Delchier JC, Celiñski K, Giguère M, et al. (2011) Helicobacter pylori eradication with a capsule containing bismuth subcitrate potassium, metronidazole and tetracycline given with omeprazole versus clarithromycin-based triple therapy: a randomised, open-label, non-inferiority, phase 3 trial. Lancet 377: 905-913.

  34. Hsu PI, Wu DC, Wu JY, Graham DY (2011) Modified sequential Helicobacter pylori therapy: Proton Pump Inhibitor and amoxicillin for 14 days with clarithromycin and metronidazole added as a quadruple (hybrid) therapy for the final seven days. Helicobacter 16: 139-145.

  35. Zhebrun AB, Alexandrova VA, Goncharova LB, Tkachenko EI (2002) Diagnostic , prophylaxis and treatment of H. pylori-associated diseases: recommendation for physicians. St-Petersburg 15-18.

  36. Megraud F (2007) Helicobacter pylori and antibiotic resistance. Gut 56: 1502.

  37. O'Connor A, Vaira D, Gisbert JP, O'Morain C (2014) Treatment of Helicobacter pylori infection 2014. Helicobacter 19 Suppl 1: 38-45.

  38. Mahachai V. Ratanachu-ek T, Myint T, Pittayanon R, Yamaoka Y, Uchida T, et al. (2014) Antibiotic resistant pattern of Helicobacter pylori infection in the south and southeast Asian countries. Helicobacter 1: 146.

  39. Alarcon T, Saez S, Merchan B, Rubio B, Gomez de Olea F, et al. (2014) Antimicrobial resistance in Helicobacter pylori and its relationship with consumption of antibiotics. Helicobacter 1: 146.

  40. Martinez-Gonzalez B, Papadakos K, Psarrakos P, Roma E, Panayiotou J, Sgouras DN, Mentis AF. High rate of calrithromycin resistance in Helicobacter pylori clinical isolates obtained from children in greece: a retrospective study, 2000–2013. Helicobacter 1: 148.

  41. Martins GM, Lima KS, Cota BDCV, Sanches BSV, Moretzsohn LD, Et al. (2014) Molecular detection of clarithromycin and fluoroquinolones resistance in Helicobacter pylori infection, directly applied to gastric biopsies, in a brazilian urban population. Helicobacter 1: 150.

  42. Mandkhai B, Munkhdelger Y, Bira N, Sarantuya J (2014) Drug resistant H. pylori strains isolated in Mongolia. Helicobacter 1: 151.

  43. Siavoshi F, Hosseini F, Shahreza S, Khalili samani S, Afshar B (2014) Increase in the resistance rates of H. pylori isolates after 5 years. Helicobacter 1: 153.

  44. Camargo MC, García A, Riquelme A, Otero W, Camargo CA, et al. (2014) The problem of Helicobacter pylori resistance to antibiotics: a systematic review in Latin America. Am J Gastroenterol 109: 485-495.

  45. Hsiang J, Selvaratnam S, Taylor S, Yeoh J, Tan YM, et al. (2013) Increasing primary antibiotic resistance and ethnic differences in eradication rates of Helicobacter pylori infection in New Zealand–a new look at an old enemy. N Z Med J 126: 64-76.

  46. An B, Moon BS, Kim H, Lim HC, Lee YC, et al. (2013) Antibiotic resistance in Helicobacter pylori strains and its effect on H. pylori eradication rates in a single center in Korea. Ann Lab Med 33: 415-419.

  47. Lee JW, Kim N, Kim JM, Nam RH, Chang H, et al. (2013) Prevalence of primary and secondary antimicrobial resistance of Helicobacter pylori in Korea from 2003 through 2012. Helicobacter 18: 206-214.

  48. Vekens K, Vandebosch S, De Bel A, Urbain D, Mana F (2013) Primary antimicrobial resistance of Helicobacter pylori in Belgium. Acta Clin Belg 68: 183-187.

  49. Selgrad M, Meissle J, Bornschein J, Kandulski A, Langner C, et al. (2013) Antibiotic susceptibility of Helicobacter pylori in central Germany and its relationship with the number of eradication therapies. Eur J Gastroenterol Hepatol 25: 1257-1260.

  50. O’Connor A, Taneike I, Nami A, Fitzgerald N, Ryan B, et al. (2013) Helicobacter pylori resistance rates for levofloxacin, tetracycline and rifabutin among Irish isolates at a reference centre. Ir J Med Sci 182: 693-635.

  51. Su P, Li Y, Li H, Zhang J, Lin L, et al. (2013) Antibiotic resistance of Helicobacter pylori isolated in the Southeast Coastal Region of China. Helicobacter 18: 274-279.

  52. De Francesco V, Giorgio F, Hassan C, Manes G, Vannella L, et al. (2010) Worldwide H. pylori antibiotic resistance: a systematic review. J Gastrointestin Liver Dis 19: 409-414.

  53. Uspenskiy YP, Suvorov AN, Baryshnikova NV (2011) Helicobacter pylori infection in clinical practice. St-Petersburg: InformMed 462-554.

  54. Vallve M, Vergara M, Gisbert JP, Calvet X (2002) Single vs. double dose of a proton pump inhibitor in triple therapy for Helicobacter pylori eradication: a meta-analysis. Aliment Pharmacol Ther 16: 1149-1156.

  55. Wang C, Ma MH, Chou HC, Yen ZS, Yang CW, et al. (2010) High-dose vs non-high-dose proton pump inhibitors after endoscopic treatment in patients with bleeding peptic ulcer: a systematic review and meta-analysis of randomized controlled trials. Arch Intern Med 170: 751-758.

  56. Paoluzi P, Iacopini F, Crispino P, Nardi F, Bella A, et al. (2006) 2-week triple therapy for Helicobacter pylori infection is better than 1-week in clinical practice: a large prospective single-center randomized study. Helicobacter 11: 562-568.

  57. Katicic M, Ticak M, Prskalo M. Eradication of Helicobacter pylori infection with two triple therapy regimes of 7, 10, and 14 days: four year experience. Helicobacter 11: 386.

  58. Calvet X, Garcia N, Lopez T, Gisbert JP, Gené E, et al. (2000) A meta-analysis of short versus long therapy with a proton pump inhibitor, clarithromycin and either metronidazole or amoxicillin for treating Helicobacter pylori infection. Aliment Pharmacol Ther 14: 603–609.

  59. Simanenkov VI, Tkachenko EI, Zakharova NV (2008) Protocols of diagnostic and treatment of H. pylori infection. St-Petersburg 3-32.

  60. Di Mario F, Cavallaro LG, Scarpignato C (2006) 'Rescue' therapies for the management of Helicobacter pylori infection. Dig Dis 24: 113-130.

  61. Gisbert JP, Romano M, Molina-Infante J, Lucendo AJ, Medina E, et al. (2015) Two-week, high-dose proton pump inhibitor, moxifloxacin triple Helicobacter pylori therapy after failure of standard triple or non-bismuth quadruple treatments. Dig Liver Dis 47: 108-113.

  62. Furuta T, Sugimoto M, Kodaira C, Nishino M, Yamade M, et al. (2014) Sitafloxacin-based third-line rescue regimens for Helicobacter pylori infection in Japan. J Gastroenterol Hepatol 29: 487-493.

  63. Lazebnik LB, Bordin DS (2013) Management of patients with Helicobacter pylori-related illness in the ordinary clinical practice. Intermediate results of monitoring program. Eksp Klin Gastroenterol 5: 93-101.

  64. Zakharova NV (2004) Evolution of view on problem of eradication of H. pylori. Consilium medicum 6: 9-11.

  65. Uspenskiy YP, Baryshnikova NV (2012) Usage of nitrofurans in eradication schemes of first-line treatment. Russiam Medical Journal 25: 1-4.

  66. Gasbarrini A, Lauritano EC, Nista EC, Candelli M, Gabrielli M, et al. (2006) Rifaximin-based regimens for eradication of Helicobacter pylori: a pilot study. Dig Dis 24: 195-200.

  67. Ciccaglione AF, Cellini L, Grossi L, Manzoli L, Marzio L (2015) A Triple and Quadruple Therapy with Doxycycline and Bismuth for First-Line Treatment of Helicobacter pylori Infection: A Pilot Study. Helicobacter 20: 390-396.

  68. Olokoba AB, Obateru OA, Bojuwoye MO (2013) Helicobacter pylori eradication therapy: A review of current trends. Niger Med J 54: 1-4.

  69. Parfenov AI, Ruchkina IN (2008) Irritable bowel disease (recommendations for physicians). Moscow, 3-34.

  70. Lazebnic LB, Tkachenko EI, Abdulkhakov RA, Bordin DS, Grinevich VB, et al. (2013) Standard of diagnostic and treatment of acid-related diseases and H. pylori-associated diseases 1: 20.

  71. Malfertheiner P (2010) Infection: Bismuth improves PPI-based triple therapy for H. pylori eradication. Nat Rev Gastroenterol Hepatol 7: 538-539.

  72. Ciccaglione AF, Cellini L, Grossi L, Marzio L (2012) Quadruple therapy with moxifloxacin and bismuth for first-line treatment of Helicobacter pylori. World J Gastroenterol 18: 4386-4390.

  73. Ghadir MR, Shafaghi A, Iranikhah A, Pakdin A, Joukar F, et al. (2011) Furazolidone, amoxicillin and omeprazole with or without bismuth for eradication of Helicobacter pylori in peptic ulcer disease. Turk J Gastroenterol 22: 1-5.

  74. Farkas R, Pronai L, Tulassay Z, Selmeci L (2005) Relationship between eradication of Helicobacter pylori and gastric mucosal superoxide dismutase activity. Anticancer Res 25: 4763-4767.

  75. Aruin LI, Kononov AV, MozgovoĬ SI (2009) [International classification of chronic gastritis: what should be taken and what is in doubt]. Arkh Patol 71: 11-18.

  76. Vakil N, Vaira D (2008) Sequential therapy for Helicobacter pylori: time to consider making the switch? JAMA 300: 1346-1347.

  77. O'Connor A, Gisbert J, O'Morain C (2009) Treatment of Helicobacter pylori infection. Helicobacter 14 Suppl 1: 46-51.

  78. Kim JS, Park SM, Kim BW (2015) Sequential or concomitant therapy for eradication of Helicobacter pylori infection: A systematic review and meta-analysis. J Gastroenterol Hepatol 30: 1338-1345.

  79. Kim SY, Park DK, Kwon KA, Kim KO, Kim YJ, et al. (2014) [Ten day concomitant therapy is superior to ten day sequential therapy for Helicobacter pylori eradication]. Korean J Gastroenterol 64: 260-267.

  80. Lee HJ, Kim JI, Lee JS, Jun EJ, Oh JH, et al. (2015). Concomitant therapy achieved the best eradication rate for Helicobacter pylori among various treatment strategies. World J Gastroenterol 21: 351-359.

  81. Metanat HA, Valizadeh SM, Fakheri H, Maleki I, Taghvaei T, et al. (2015) Comparison Between 10- and 14-Day Hybrid Regimens for Helicobacter pylori Eradication: A Randomized Clinical Trial. Helicobacter 20: 299-304.

  82. Cuadrado-Lavín A, Salcines-Caviedes JR, Diaz-Perez A, Carrascosa MF, Ochagavía M, et al. (2015) First-line eradication rates are comparing two shortened non-bismuth quadruple regimens against Helicobacter pylori: an open-label, randomized, multicentre clinical trial. J Antimicrob Chemother.

  83. He L, Deng T, Luo H (2015) Meta-analysis of sequential, concomitant and hybrid therapy for Helicobacter pylori eradication. Intern Med 54: 703-710.

  84. Wang B, Wang YH, Lv ZF, Xiong HF, Wang H, et al. (2015) Review: efficacy and safety of hybrid therapy for Helicobacter pylori infection: a systematic review and meta-analysis. Helicobacter 20: 79-88.

  85. Felley C, Michetti P (2003) Probiotics and Helicobacter pylori. Best Pract Res Clin Gastroenterol 17: 785-791.

  86. Szajewska H, Horvath A, Piwowarczyk A (2010) Meta-analysis: the effects of Saccharomyces boulardii supplementation on Helicobacter pylori eradication rates and side effects during treatment. Aliment Pharmacol Ther 32: 1069-1079.

  87. Molina-Infante J, Gisbert JP (2013) Probiotics for Helicobacter pylori eradication therapy: not ready for prime time. Rev Esp Enferm Dig 105: 441-444.

  88. Zhu R, Chen K, Zheng YY, Zhang HW, Wang JS, et al. (2014) Meta-analysis of the efficacy of probiotics in Helicobacter pylori eradication therapy. World J Gastroenterol 20: 18013-18021.

  89. Dang Y, Reinhardt JD, Zhou X, Zhang G (2014) The effect of probiotics supplementation on Helicobacter pylori eradication rates and side effects during eradication therapy: a meta-analysis. PLoS One 9: e111030.

  90. Tong JL, Ran ZH, Shen J, Zhang CX, Xiao SD (2007) Meta-analysis: The effect of supplementation with probiotics on eradication rates and adverse events during Helicobacter pylori eradication therapy. Aliment Pharmacol Ther 25: 155-168.

  91. Wang ZH, Gao QY, Fang JY (2013) A meta-analysis of the efficacy and safety of Lactobacillus-containing and Bifidobacterium-containing probiotic compound preparation in Helicobacter pylori eradication therapy. J Clin Gastroenterol 47: 25-32.

  92. Sachdeva A, Nagpal J (2009) Meta-analysis: efficacy of bovine lactoferrin in Helicobacter pylori eradication. Aliment Pharmacol Ther 29: 720-730.

  93. Zou J, Dong J, Yu XF (2009) Meta-analysis: the effect of supplementation with lactoferrin on eradication rates and adverse events during Helicobacter pylori eradication therapy. Helicobacter 14: 119-127.

  94. Sachdeva A, Nagpal J (2009) Effect of fermented milk-based probiotic preparations on Helicobacter pylori eradication: A systematic review and meta-analysis of randomized-controlled trials. Eur J Gastroenterol Hepatol 21: 45-53.

  95. Zou J, Dong J, Yu X (2009) Meta-analysis: Lactobacillus containing quadruple therapy versus standard triple first-line therapy for Helicobacter pylori eradication. Helicobacter 14: 97-107.

  96. Dinleyici EC, Kara A, Ozen M, Vandenplas Y (2014) Saccharomyces boulardii CNCM I-745 in different clinical conditions. Expert Opin Biol Ther 14: 1593-1609.

  97. Srinarong C, Siramolpiwat S, Wongcha-um A, Mahachai V, Vilaichone RK (2014) Improved eradication rate of standard triple therapy by adding bismuth and probiotic supplement for Helicobacter pylori treatment in Thailand. Asian Pac J Cancer Prev 15: 9909-9913.

  98. Akcam M, Koca T, Salman H, Karahan N (2015) The effects of probiotics on treatment of Helicobacter pylori eradication in children. Saudi Med J 36: 286-290.

  99. Lv Z, Wang B, Zhou X, Wang F, Xie Y, et al. (2015) Efficacy and safety of probiotics as adjuvant agents for Helicobacter pylori infection: A meta-analysis. Exp Ther Med 9: 707-716.

  100. Kozlova DI (2004) Intestinal microbiocenosis and H. pylori-associated gastritis after eradication and sinbiotic therapy. St-Petersburg, 3-21.

  101. Baryshnikova NV, Uspenskiy YP, Suvorov AN, Suvorova MA, Tkachenko EI (2011) Using monotherapy of probiotics as an alternative method in treatment of Helicobacter pylori infection. Slovakia, 13-14.

  102. Canducci F, Cremonini F, Armuzzi A, Di Caro S, Gabrielli M, et al. (2002) Probiotics and Helicobacter pylori eradication. Dig Liver Dis 34 Suppl 2: S81-83.

  103. Suvorov AN, Simanenkov VI, Zakharova NV, Baryshnikova NV (2013) Efficacy of Enterococcus faecium L3 in therapy of Helicobacter pylori infection. Proceeding of the International Scientific conference on bacteriocins and antimicrobial peptides. Slovakia, 28.

  104. Russo F, Linsalata M, Orlando A (2014) Probiotics against neoplastic transformation of gastric mucosa: effects on cell proliferation and polyamine metabolism. World J Gastroenterol 20: 13258-13272.

  105. Vilaichone RK, Mahachai V, Tumwasorn S, Nunthapisud P, Kullavanijaya P (2002) Inhibitory effect of Lactobacillus acidophilus on Helicobacter pylori in peptic ulcer patients: in vitro study. J Med Assoc Thai 85 Suppl 1: S79-84.

  106. Baryshnikova NV, Uspenskiy YP, Zhebrun AB, Svarval AV, Ferman RS, ET AL. (2014) Efficacy of different probiotics as antihelicobacter medications in vitro. Helicobacter 19: 156.

  107. Shenderov BA (2013) Metabiotics: novel idea or natural development of probiotic conception. Microb Ecol Health Dis 24.

  108. Holz C, Busjahn A, Mehling H, Arya S, Boettner M, et al. (2014) Significant Reduction in Helicobacter pylori Load in Humans with Non-viable Lactobacillus reuteri DSM17648: A Pilot Study. Probiotics Antimicrob Proteins 7: 91-100.

  109. Mehling H, Busjahn A (2013) Non-viable Lactobacillus reuteri DSMZ 17648 (Pylopassâ„¢) as a new approach to Helicobacter pylori control in humans. Nutrients 5: 3062-3073.

  110. Blinkova LP, Al'tshuler ML, Dorofeeva ES, Gorobets OB (2007) Molecular basis of bacteriocins production and activity. Zh Mikrobiol Epidemiol Immunobiol : 97-104.

  111. Kim TS, Hur JW, Yu MA, Cheigh CI, Kim KN, et al. (2003) Antagonism of Helicobacter pylori by bacteriocins of lactic acid bacteria. J Food Prot 66: 3-12.

  112. Shi T, Liu WZ, Gao F, Shi GY, Xiao SD (2005) Intranasal CpG-oligodeoxynucleotide is a potent adjuvant of vaccine against Helicobacter pylori, and T helper 1 type response and interferon-gamma correlate with the protection. Helicobacter 10: 71-79.

  113. Hatzifoti C, Roussel Y, Harris AG, Wren BW, Morrow JW, et al. (2006) Mucosal immunization with a urease B DNA vaccine induces innate and cellular immune responses against Helicobacter pylori. Helicobacter 2: 113–122.

  114. Ilver D, Arnqvist A, Ogren J, Frick IM, Kersulyte D, et al. (1998) Helicobacter pylori adhesin binding fucosylated histo-blood group antigens revealed by retagging. Science 279: 373-377.

  115. Graham DY, Opekun AR, Osato MS, El-Zimaity HM, Lee CK, et al. (2004) Challenge model for Helicobacter pylori infection in human volunteers. Gut 53: 1235-1243.

  116. Malfertheiner P, Schultze V, Rosenkranz B, Kaufmann SH, Ulrichs T, et al. (2008) and immunogenicity of an intramuscular Helicobacter pylori vaccine in noninfected volunteers: a phase I study. Gastroenterology 135: 787-795.

  117. Xu C, Li ZS, Du YQ, Gong YF, Yang H, et al. (2007) Construction of recombinant attenuated Salmonella typhimurium DNA vaccine expressing H pylori ureB and IL-2. World J Gastroenterol 13: 939-944.

  118. Stasilojc M, Hinc K, Peszynska-Sularz G, Obuchowski M, Iwanicki A (2015) Recombinant Bacillus subtilis Spores Elicit Th1/Th17-Polarized Immune Response in a Murine Model of Helicobacter pylori Vaccination. Mol Biotechnol 57: 685-691.

  119. Zhang HX, Qiu YY, Zhao YH, Liu XT, Liu M, et al. (2014) Immunogenicity of oral vaccination with Lactococcus lactis derived vaccine candidate antigen (UreB) of Helicobacter pylori fused with the human interleukin 2 as adjuvant. Mol Cell Probes 28: 25-30.

  120. Gu Q, Song D, Zhu M (2009) Oral vaccination of mice against Helicobacter pylori with recombinant Lactococcus lactis expressing urease subunit B. FEMS Immunol Med Microbiol 56: 197-203.

  121. Smythies LE, Novak MJ, Waites KB, Lindsey JR, Morrow CD, et al. (2005) Poliovirus replicons encoding the B subunit of Helicobacter pylori urease protects mice against H. pylori infection. Vaccine 23: 901-909.

  122. D'Elios MM, Andersen LP (2007) Helicobacter pylori inflammation, immunity, and vaccines. Helicobacter 12 Suppl 1: 15-19.

  123. Garnock-Jones KP (2015) Vonoprazan: first global approval. Drugs 75: 439-443.

  124. Inaba T, Iwamuro M, Toyokawa T, Okada H (2016) Letter: promising results of Helicobacter pylori eradication with vonoprazan-based triple therapy after failure of proton pump inhibitor-based triple therapy. Aliment Pharmacol Ther 43: 179-180.

  125. Lazebnik LB, Vasil'ev IuV, Shcherbakov PL, Khomeriki SG, Masharova AA, et al. (2010) [Helicobacter pylori: prevalence, diagnosis, treatment]. Eksp Klin Gastroenterol : 3-7.

  126. Dubtsova EA, Morozov IA, Kas'ianenko VI, Komissarenko IA (2006) [Propolis anti-Helicobacter efficacy]. Eksp Klin Gastroenterol : 94-95.

  127. Malinovskaia NK, Rapoport SI, Zhernakova NI, Rybnikova SN, Postnikova LI, et al. (2007) [Antihelicobacter effects of melatonin]. Klin Med (Mosk) 85: 40-43.

  128. Nikitin OL, Drach DA, Fadeeva IA (2010) Ozonetherapy: physiological method in prophylaxis and treatment gastro-duodenal ulcers in elder patients. Russian J Gastroenterol Hepatol Coloproctol 20: 33.

  129. Fedorov AA, Gromov AS, Sapronenok SV, Kurochkin VIu, Zhernakova ZM (2006) [Ozone therapy in gastroduodenal pathology associated with Helicobacter pylori]. Vopr Kurortol Fizioter Lech Fiz Kult : 34-37.

  130. Averianov PF, Islamova EA, Chizh AG (2005) Physical methods of therapy of gastro-intestinal tract diseases. Fundamental researches 10: 29-30.

  131. Khomeriki SG (1999) Effect of laserotherapy on Helicobacter pylori, resistance to metronidazole. In: Khomeriki SG, Khomeriki NM (eds) Helicobacter pylori:revolution in gastroenterology. Moscow, Triade, 1999: 219-223.

  132. Pavlov ON, Aleksandrov IuK (2010) [The laser chromoendoscopy eradication of Helicobacter pylori]. Eksp Klin Gastroenterol : 32-36.

  133. van Doorn LJ, Schneeberger PM, Nouhan N, Plaisier AP, Quint WG, et al. (2000) Importance of Helicobacter pylori cagA and vacA status for the efficacy of antibiotic treatment. Gut 46: 321-326.

  134. Höcker M, Hohenberger P (2003) Helicobacter pylori virulence factors--one part of a big picture. Lancet 362: 1231-1233.

  135. Chukov SZ, Morozov IA, Pasechnikov VD (2002) [Clinico-morphological and molecular-genetic correlations in the stomach pathology associated with Helicobacter pylori]. Arkh Patol 64: 37-40.

  136. Figura N, Guglielmetti P, Rossolini A, Barberi A, Cusi G, et al. (1989) Cytotoxin production by Campylobacter pylori strains isolated from patients with peptic ulcers and from patients with chronic gastritis only. J Clin Microbiol 27: 225-226.

  137. Yoshimura HH, Evans DG, Graham DY (1990) H. pylori strains from duodenal ulcer patients differ at the genomic level from those from patients with simple gastritis. Rev. Esp. Enf. Digest 78: 6.

  138. Kuo CH, Lu CY, Shih HY, Liu CJ, Wu MC, et al. (2014) CYP2C19 polymorphism influences Helicobacter pylori eradication. World J Gastroenterol 20: 16029-16036.

  139. Zhao F, Wang J, Yang Y, Wang X, Shi R, et al. (2008) Effect of CYP2C19 genetic polymorphisms on the efficacy of proton pump inhibitor-based triple therapy for Helicobacter pylori eradication: a meta-analysis. Helicobacter 13: 532-541.

  140. Maev IV, Momynaliev KT, Oganesian TS, Govorun VM, KucheriawyĬ IuA, et al. (2009) [Polymorphism of cytochrome P-450 gene in the light of Helicobacter pylori eradication efficiency]. Klin Med (Mosk) 87: 35-39.

  141. Ishida Y, Goto Y, Kondo T, Kurata M, Nishio K, et al. (2006) Eradication rate of Helicobacter pylori according to genotypes of CYP2C19, IL-1B, and TNF-A. Int J Med Sci 3: 135-140.

  142. Sugimoto M, Furuta T, Yamaoka Y (2009) Influence of inflammatory cytokine polymorphisms on eradication rates of Helicobacter pylori. J Gastroenterol Hepatol 24: 1725-1732.

  143. Leonard GD, Fojo T, Bates SE (2003) The role of ABC transporters in clinical practice. Oncologist 8: 411-424.

  144. Hoffmeyer S, Burk O, von Richter O, Arnold HP, Brockmöller J, et al. (2000) Functional polymorphisms of the human multidrug-resistance gene: multiple sequence variations and correlation of one allele with P-glycoprotein expression and activity in vivo. Proc Natl Acad Sci USA 97: 3473–3478.

  145. Siegmund W, Ludwig K, Giessmann T, Dazert P, Schroeder E, et al. (2002) The effects of the human MDR1 genotype on the expression of duodenal P-glycoprotein and disposition of the probe drug talinolol. Clin Pharmacol Ther 72: 572-583.

International Journal of Anesthetics and Anesthesiology (ISSN: 2377-4630)
International Journal of Blood Research and Disorders   (ISSN: 2469-5696)
International Journal of Brain Disorders and Treatment (ISSN: 2469-5866)
International Journal of Cancer and Clinical Research (ISSN: 2378-3419)
International Journal of Clinical Cardiology (ISSN: 2469-5696)
Journal of Clinical Gastroenterology and Treatment (ISSN: 2469-584X)
Clinical Medical Reviews and Case Reports (ISSN: 2378-3656)
Journal of Dermatology Research and Therapy (ISSN: 2469-5750)
International Journal of Diabetes and Clinical Research (ISSN: 2377-3634)
Journal of Family Medicine and Disease Prevention (ISSN: 2469-5793)
Journal of Genetics and Genome Research (ISSN: 2378-3648)
Journal of Geriatric Medicine and Gerontology (ISSN: 2469-5858)
International Journal of Immunology and Immunotherapy (ISSN: 2378-3672)
International Journal of Medical Nano Research (ISSN: 2378-3664)
International Journal of Neurology and Neurotherapy (ISSN: 2378-3001)
International Archives of Nursing and Health Care (ISSN: 2469-5823)
International Journal of Ophthalmology and Clinical Research (ISSN: 2378-346X)
International Journal of Oral and Dental Health (ISSN: 2469-5734)
International Journal of Pathology and Clinical Research (ISSN: 2469-5807)
International Journal of Pediatric Research (ISSN: 2469-5769)
International Journal of Respiratory and Pulmonary Medicine (ISSN: 2378-3516)
Journal of Rheumatic Diseases and Treatment (ISSN: 2469-5726)
International Journal of Sports and Exercise Medicine (ISSN: 2469-5718)
International Journal of Stem Cell Research & Therapy (ISSN: 2469-570X)
International Journal of Surgery Research and Practice (ISSN: 2378-3397)
Trauma Cases and Reviews (ISSN: 2469-5777)
International Archives of Urology and Complications (ISSN: 2469-5742)
International Journal of Virology and AIDS (ISSN: 2469-567X)
More Journals

Contact Us

ClinMed International Library | Science Resource Online LLC
3511 Silverside Road, Suite 105, Wilmington, DE 19810, USA
Email: contact@clinmedlib.org
 

Feedback

Get Email alerts
 
Creative Commons License
Open Access
by ClinMed International Library is licensed under a Creative Commons Attribution 4.0 International License based on a work at https://clinmedjournals.org/.
Copyright © 2017 ClinMed International Library. All Rights Reserved.