Skip to main content

REVIEW article

Front. Cell. Infect. Microbiol., 25 November 2022
Sec. Microbiome in Health and Disease
This article is part of the Research Topic Helicobacter pylori and Its Mechanisms of Antibiotic Survival View all 5 articles

Current and future perspectives for Helicobacter pylori treatment and management: From antibiotics to probiotics

Bing LiangBing Liang1Yang YuanYang Yuan1Xiao-Jin PengXiao-Jin Peng1Xin-Lin LiuXin-Lin Liu1Xiao-Kun HuXiao-Kun Hu2Dong-Ming Xing,*Dong-Ming Xing1,3*
  • 1Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
  • 2Intervention Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
  • 3School of Life Sciences, Tsinghua University, Beijing, China

Helicobacter pylori (H. pylori) is a Gram-negative anaerobic bacterium that colonizes the human stomach and is the leading cause of gastric diseases such as chronic gastritis and peptic ulcers, as well as the most definite and controllable risk factor for the development of gastric cancer. Currently, the regimen for H. pylori eradication has changed from triple to quadruple, the course of treatment has been extended, and the type and dose of antibiotics have been adjusted, with limited improvement in efficacy but gradually increasing side effects and repeated treatment failures in an increasing number of patients. In recent years, probiotics have become one of the most important tools for supporting intestinal health and immunity. Numerous in vitro studies, animal studies, and clinical observations have demonstrated that probiotics have the advantage of reducing side effects and increasing eradication rates in adjuvant anti-H. pylori therapy and are a valuable supplement to conventional therapy. However, many different types of probiotics are used as adjuncts against H. pylori, in various combinations, with different doses and timing, and the quality of clinical studies varies, making it difficult to standardize the results. In this paper, we focus on the risk, status, prevention, control, and treatment of H. pylori infection and review international consensus guidelines. We also summarize the available scientific evidence on using Limosilactobacillus reuteri (L. reuteri) as a critical probiotic for H. pylori treatment and discuss its clinical research and application from an evidence-based perspective.

Introduction

Helicobacter pylori (H. pylori) is a Gram-negative obligate anaerobic bacteria colonized in the human stomach. H. pylori is the most common infection in the world, infecting around 50% of the world’s population. It causes peptic ulcers and gastric cancer, as well as everyday digestive discomfort. Many cases of H. pylori are asymptomatic, but all individuals with H. pylori infection have degrees of gastritis, which can progress to severe symptoms over time. Eradication treatment of H. pylori infection not only improves the gastrointestinal disease associated with it but also reduces the risk of gastric cancer. The current treatment guideline is to eradicate H. pylori using a combination of two antibiotics and a proton pump inhibitor known as triple therapy (Chey et al., 2007; Malfertheiner et al., 2007; Fock et al., 2009). In some cases, a fourth drug, the anti-parasitic compound bismuth, is used, wherein we talk about quadruple therapy. The success of medical eradication ranges from 70% to 95%. The rates, however, have been declining due to increased antibiotic resistance. Furthermore, with the rise of gut microbiome research, the potential side effects of antibiotic therapy (particularly repeated long-term antibiotic use) on gut microecology have gained widespread attention (Mohsen et al., 2020). Medical H. pylori therapies cause severe side effects, which reduce treatment compliance. They also drive antibiotic resistance and cannot be taken over the long term. Hence, there is a need for effective natural solutions to control H. pylori. Researchers have begun to look for other complementary and alternative therapies to address these concerns and challenges. Probiotics are microorganisms that are beneficial to human health. A large body of basic and clinical research focuses on the different health benefits of probiotics, including their use as an adjunct to H. pylori eradication therapy. In a series of in vitro and in vivo studies, L. reuteri DSM 17648 has been shown to specifically bind to H. pylori in the gastric environment to form co-polymers that interfere with H. pylori adhesion to the gastric mucosa and facilitate its elimination, thereby reducing the H. pylori load in the stomach. Clinical trials in multiple countries have shown that L. reuteri DSM 17648 reduces H. pylori load, improves gastrointestinal discomfort, and reduces the side effects of antibiotic therapy in both adults and pediatric subjects. This article focuses on the current status, risks, and treatment strategies of H. pylori infection and reviews the relevant research progress and major consensus guidelines. Current scientific evidence for the use of L. reuteri DSM 17648 in treating H. pylori is summarized, and its clinical research and application are discussed from an evidence-based perspective.

The current status and risks of H. pylori infection

H. pylori is a common pathogenic bacterium in the stomach and is closely associated with the development of many gastrointestinal diseases (Suerbaum and Michetti, 2002; Malfertheiner et al., 2017) (Figure 1). About 25-30% of infected individuals can develop gastrointestinal diseases such as dyspepsia, gastritis, peptic ulcer, and gastric cancer (Liu et al., 2018). The gut microbiota is extremely dynamic and could be influenced by various factors, including host lifestyle, long-term proton pump inhibitor (PPI) use, antibiotic therapy, and H. pylori infection (Schmidt et al., 2018; Zhang et al., 2019). Thus, both infection and eradication of H. pylori and its interaction with the gut microbiota can alter the microecological balance, thereby affecting the onset and progression of associated diseases (Chen et al., 2022; Fakharian et al., 2022). After infection, H. pylori can adapt to the stomach’s harsh acidic environment (Jones et al., 2018; Wen et al., 2018) and interact with host cell receptors to colonize the gastric mucosa (Gonciarz et al., 2019; Hamedi Asl et al., 2019; Saenz et al., 2019), form biofilms (Hathroubi et al., 2018; Ronci et al., 2019), interfere with host metabolic pathways (Hathroubi et al., 2018; Matsunaga et al., 2018), induce neuroimmune crosstalk (Sticlaru et al., 2018), and down-regulate gastric barrier homeostasis (Datta et al., 2018; Liu et al., 2018; Liu et al., 2018; Yaseen and Audette, 2018). More importantly, both H. pylori infection and eradication therapy could lead to gut microbiota disturbance, resulting in ecological dysbiosis, promoting gastric carcinogenesis and tumorigenesis (Kienesberger et al., 2016; Ansari and Yamaoka, 2017; Li et al., 2017; Llorca et al., 2017; Guo et al., 2020).

FIGURE 1
www.frontiersin.org

Figure 1 Gastrointestinal disease outcomes of H. pylori infection.

Fifth Chinese National Consensus Report on the Management of H. pylori infection specifically states that peptic ulcers occur in 15-20% of H. pylori-infected patients, dyspepsia in 5-10%, and gastric malignancy in about 1% (Table 1). Once infected, H. pylori infection is difficult to cure on its own without treatment. Due to the high prevalence of H. pylori infection worldwide, its related diseases pose a heavy disease burden for human health. H. pylori infection is an important cause of gastric cancer (Smyth et al., 2020). Epidemiological surveys have shown that H. pylori infection increases the risk of gastric cancer by 4-6 times (Forman et al., 1994). The currently recognized model of gastric cancer disease suggests that H. pylori infection drives the progression of the normal gastric mucosa to chronic active gastritis, atrophic gastritis, intestinal metaplasia, dysplasia, and gastric cancer (Correa, 1992; O'connor et al., 2017) (Figure 2). Therefore, H. pylori eradication is equivalent to the removal of an initiating factor for gastric cancer development. A large meta-analysis also confirmed that H. pylori eradication treatment effectively reduced the relative risk of gastric cancer by 46% in healthy infected individuals (Ford et al., 2020). Overall, the prevalence of H. pylori infection has decreased in developed and some developing countries, which may be mainly related to improving living standards and hygiene conditions. Notably, the overall global prevalence of H. pylori infection is still higher than 40%, and according to the predictions of the meta-analysis, the number of people infected with H. pylori worldwide could be as high as 4.4 billion (Hooi et al., 2017). Moreover, there is heterogeneity across studies, with some regional studies unable to express overall prevalence. According to this study, there is an overall global annual recurrence rate of H. pylori infection (negative test results after eradication therapy and reappearance of positive H. pylori at follow-up) of 4.3%. The annual rate of re-infection (re-infection after eradication) is 3.1%, and the annual re-ignition rate (mainly due to unsuccessful eradication programs) is 2.2%. The recurrence rate is mainly related to the level of economic and social development and health conditions. Thus H. pylori-associated diseases continue to pose a significant disease burden on human health.

TABLE 1
www.frontiersin.org

Table 1 Incidence of H. pylori infection-associated disease in infected individuals and the proportion attributable to infection.

FIGURE 2
www.frontiersin.org

Figure 2 Disease progression pattern of H. pylori infection promoting gastric cancer: chronic active gastritis occurs after infection of normal gastric mucosa with H. pylori, through progressive development of atrophic gastritis, intestinal chemosis, heterogeneous hyperplasia, and finally gastric cancer.

Strategies for prevention, control, and treatment of H. pylori infection and challenges

According to the Kyoto Consensus and the Maastricht V Consensus, the causal relationship between H. pylori infection and chronic active gastritis is in line with the Koch principle. It is an infectious disease transmitted from person to person (Sugano et al., 2015; Malfertheiner et al., 2017). H. pylori transmission necessitates three primary conditions: the source of infection, the route of transmission, and the susceptible population. According to the Kyoto Consensus, all patients with H. pylori infection require treatment unless countervailing factors exist (Sugano et al., 2015). According to the Taipei Consensus, the greatest benefit is from H. pylori eradication in young adults, which are marital and parenting, and eradicating H. pylori in young adults can cure H. pylori-associated gastritis, reduce the risk of gastric cancer, and eliminate household transmission (Liou et al., 2020). The second is to cut off the transmission route. H. pylori is primarily transmitted orally, including oral-oral transmission, fecal-oral transmission, shared appliance transmission and water source transmission (NCMRCFDD (Shanghai), 2021), with oral-oral transmission through saliva being the most common cause of household cluster infections (chewing and feeding) (Yokota et al., 2015). People of all ages are generally susceptible to H. pylori, with infants and children being the most vulnerable, with most patients infected in childhood, especially before the age of 12 (Sugano et al., 2015; Chey et al., 2017).

Presently, international consensus emphasizes the importance of eradication treatment for people infected with H. pylori. Drugs for the eradication of H. pylori include antibiotics and proton pump inhibitors (PPIs), where antibiotics work to kill H. pylori directly, while PPIs work to inhibit gastric acid secretion and raise the pH level in the stomach to create the right environment for the antibiotics to exert their bactericidal effect. A single drug cannot eradicate H. pylori, and a combination treatment regimen must be used. The main clinical protocols are triple and quadruple therapy and the resulting sequential, concomitant, and mixed therapies. The leading international consensus opinions on H. pylori eradication treatment are the Toronto Consensus on the Treatment of Adult H. pylori Infection (Toronto Consensus) (Fallone et al., 2016), American College of Gastroenterology Consensus on the Treatment of H. pylori Infection (ACG Consensus) (Chey et al., 2017), Maastricht V Consensus (Malfertheiner et al., 2017) and Fifth Chinese National Consensus (Liu et al., 2018), whose primary treatment regimens are summarized and compared in Table 2.

TABLE 2
www.frontiersin.org

Table 2 International consensus opinion on the recommended primary treatment regimen for H. pylori infection.

Triple therapy

A combination of one PPI with two antibiotics is a triple therapy. The standard triple regimen based on clarithromycin was established in 1996 (Lind et al., 1996). It can be taken for 7 days with omeprazole, amoxicillin, or omeprazole and metronidazole, with clarithromycin doses of 500 mg and 250 mg, respectively. This regimen was the first-line regimen for H. pylori eradication at the time because of its low drug intake, short course of treatment, high efficacy, and low incidence of side effects. Later, scholars expanded the triple regimen with levofloxacin or metronidazole, achieving high eradication rates. However, with the increasing rate of antibiotic resistance of H. pylori over the years, the eradication rate of these drug-based triple regimens has been below or well below 80%, and the eradication rate is unsatisfactory even if the regimen is extended to 10 or even 14 days (Malfertheiner et al., 2011; Malfertheiner et al., 2012).

Bismuth quadruple therapy

The classic bismuth quadruple regimen, which dates back to 1995 and consists of PPI, bismuth, tetracycline, and metronidazole, was established before the clarithromycin triple regimen (Graham and Lee, 2015). As clarithromycin triplet was the first-line regimen at that time, bismuth quadruplet was used only as a remedial treatment. As the rate of H. pylori resistance to clarithromycin increased, the efficacy of the clarithromycin triplet regimen declined, and bismuth quadruple therapy was relegated to first-line treatment. Bismuth increases the eradication rate of H. pylori resistant strains by 30% to 40% (Dore et al., 2016). Despite the high resistance rates of clarithromycin, metronidazole, and levofloxacin, adding bismuth to triple regimens containing these agents has resulted in satisfactory efficacy (Zhang et al., 2015). The Maastricht V, Toronto, and ACG consensus recommend bismuth quadruple therapy as the first-line treatment option due to the high eradication rate and the fact that bismuth is not easily developed drug resistance and has high safety in short-term application.

Non-Bismuth Quadruple therapy

Depending on the mode of administration, a non-Bismuth Quadruple regimen can be divided into sequential therapy (PPI + amoxicillin for the first 5 or 7 days and PPI + clarithromycin + metronidazole for the second 5 or 7 days), concomitant therapy (4 drugs for 10 or 14 days), and mixed therapy (same as sequential therapy for the first 5 or 7 days and concomitant therapy for the second 5 or 7 days). Of these regimens, concomitant therapy with 3 antibiotics is the most effective in overcoming antibiotic resistance and therefore has the best relative efficacy but also has a correspondingly higher side effect profile. Because sequential therapies are vulnerable to single resistance to clarithromycin or metronidazole, they have been ruled out for adult treatment by the Maastricht V and Toronto consensus (Fallone et al., 2016; Malfertheiner et al., 2017). When both clarithromycin and metronidazole become resistant, non-bismuth quadruple therapy effectively becomes PPI plus amoxicillin two-component therapy, and eradication rates for sequential, mixed, and concomitant therapy are all reduced (Malfertheiner et al., 2017). Non-bismuth quadruple therapy with clarithromycin and metronidazole is not recommended for empirical treatment in areas with >15% dual resistance of H. pylori to clarithromycin and metronidazole, according to the Fifth Chinese National Consensus (Liu et al., 2018).

Combined Chinese and Western medicine therapy

Commonly used combined Chinese and Western medicine therapies include herbal combination Triple therapy or bismuth quadruple therapy, of which herbal combination triple therapy has the most clinical evidence. Studies have found that certain herbal combination triple therapy has comparable eradication rates to bismuth quadruple therapy at 14 days, while clinical symptom relief is superior to bismuth quadruple therapy (Yao X.J, 2018). Chinese herbal medicine combined with bismuth quadruple therapy has been used relatively rarely because of excessive drug use and minor improvement in the eradication rate. According to the Chinese Expert Consensus on Collaborative Diagnosis and Treatment of Gastritis Caused by Helicobacter pylori in Adults, Chinese herbal medicine can be combined with different aspects of bismuth quadruple remedy treatment to identify symptoms in order to improve the eradication rate of bismuth quadruple remedy treatment (Zhang and Lan, 2020). Patients with obvious GI symptoms can use Traditional Chinese Medicine (TCM) supported by evidence-based medical evidence 2 weeks before the application of bismuth quadruple therapy. Patients without obvious GI symptoms can be treated with TCM supported by evidence-based medical evidence for 2 weeks after bismuth quadruple therapy.

Other therapies

Quinolones have multiple indications. They are widely used in clinical settings and are cross-resistant to one another, so levofloxacin resistance is widespread except in a few areas. Fifth Chinese National Consensus, Maastricht V Consensus, and Toronto Consensus do not recommend levofloxacin-containing therapy for initial treatment, but it can be used as a remedial treatment option for the failure of first-line therapy (clarithromycin-based) and second-line therapy (classical bismuth quadruple regimen); there is evidence that high-dose PPI and amoxicillin in a duo regimen can achieve high eradication rates, but clinical evidence is scarce and has not been included in mainstream consensus guidelines There is evidence that the addition of certain probiotics to conventional therapies can reduce some of the side effects, but there is a lack of solid evidence to improve eradication rates.

Overall, unless there are clear drug sensitivity test results or evidence of low resistance rates, triple therapy is now not an option as a first-line treatment option. The efficacy of non-bismuth quadruple regimens is vulnerable to single or dual resistance to clarithromycin and metronidazole. Bismuth quadruple therapy, with its high eradication rate and low resistance to bismuth, is recommended by major mainstream consensus opinions as the best option for initial treatment or in areas with >15% clarithromycin resistance.

Challenges

The eradication rate of conventional H. pylori regimens is on the decline globally, and the treatment of H. pylori infection faces many challenges, including antibiotic resistance, treatment side effects, patient compliance, and re-infection. Although H. pylori antimicrobial resistance varies by geographic region, its prevalence has been increasing over time, resulting in therapy failures and low eradication rates (Flores-Trevino et al., 2018; EaY Tshibangu-Kabamba, 2021). In 2017, H. pylori were listed by the World Health Organization as one of the 20 pathogenic bacteria that pose a significant threat to human health due to drug resistance (Tacconelli et al., 2018). In 2018, a meta-analysis showed that the current H. pylori resistance rates to clarithromycin, levofloxacin, and metronidazole exceed the Maastricht V consensus recommended threshold for high resistance rates (15%) in most regions worldwide, with dual resistance rates of >15% to clarithromycin and metronidazole in some regions. In contrast, resistance rates to amoxicillin, tetracycline, and furazolidone remain low (Savoldi et al., 2018). High clarithromycin resistance has led to decreasing eradication rates of previous first-line clarithromycin-containing triple therapy (PPI/ranitidine bismuth citrate, clarithromycin, amoxicillin/metronidazole). Studies have shown that the eradication rate of clarithromycin-containing triple therapy has decreased to less than 70% (Agudo et al., 2010). In addition to causing secondary resistance to H. pylori, antibiotic therapy may also result in many side effects. In 2021, a study of 22,000 patients showed that approximately 23% of patients experienced at least one H. pylori eradication-related side effect, with taste disturbance (7%), diarrhea (7%), nausea (6%), and abdominal pain (3%) being the most common (Nyssen et al., 2021). Furthermore, to design optimized H. pylori therapy, clinicians must also consider the importance of gastric cancer regression after H. pylori eradication, and pre-treatment susceptibility tests using molecular methods could be performed (Abadi and Yamaoka, 2018).

Probiotic intervention

Due to the decreasing eradication rate of conventional therapies, some studies have begun to focus on the role of probiotics in H. pylori eradication. According to the standard definition of the Food and Agriculture Organization of the United Nations and the World Health Organization, probiotics are live microorganisms that, when ingested in sufficient quantities, are beneficial to host health (Hill et al., 2014). Numerous studies have shown that probiotics can benefit the human body in many ways, mainly in improving the health of the gastrointestinal tract (Hill et al., 2014). Current research on probiotics in H. pylori eradication treatment focuses on whether the addition of probiotics can improve the eradication rate of H. pylori; whether the addition of probiotics can reduce the incidence of side effects and alleviate symptoms in H. pylori eradication regimens; and whether the addition of probiotics can promote the restoration of microecological imbalances caused by eradication drugs. The potential mechanisms of action of probiotics to improve H. pylori infection include the following aspects (Figure 3). First, probiotics may help to enhance the barrier effect (Suez et al., 2019). The gastric acid and mucus barrier of the gastric mucosa are the first line of defense against pathogenic bacteria in the stomach. Some probiotics can upregulate tight junction protein expression, promote mucin and mucus secretion and thus mucus secretion, and enhance the barrier effect of the gastric mucosa. Second, some probiotics can secrete antimicrobial substances, such as lactic acid, short-chain fatty acids (SCFAs), hydrogen peroxide, and bacteriocins (Homan and Orel, 2015). Lactic acid and SCFAs have incomplete dissociation properties, and the undissociated forms of these organic acids can cause damage to H. pylori. The anti-H. pylori effect of lactic acid and SCFAs is also related to their inhibition of H. pylori urease activity. Some probiotics can synthesize hydrogen peroxide and bacteriocins, which also have direct antibacterial effects. Third, probiotics can interfere with H. pylori colonization (Qureshi et al., 2019; Ji and Yang, 2020). Some probiotics can interfere with the colonization of H. pylori in gastric mucosal epithelial cells by competing for adhesion sites, interfering with the adhesion process, and binding to H. pylori to form co-polymers to facilitate its excretion. In addition to these non-immune effects, some probiotics may also reduce the host inflammatory response caused by H. pylori infection (Ji and Yang, 2020). The sustained expression of inflammatory factors caused by H. pylori infection can lead to a long-term chronic inflammatory response and is an important pathological basis for the pathogenesis of H. pylori infection. Probiotics can inhibit the expression of pro-inflammatory factors and improve the inflammatory response in the stomach. Numerous clinical studies have reported the use of probiotics alone or in combination with antibiotics for H. pylori eradication. Buckley et al. showed that L. reuteri DSM 17648 effectively reduced bacterial load in the stomach and alleviated dyspeptic symptoms in H. pylori-infected patients (Buckley et al., 2018). Gotteland et al. showed that using Saccharomyces boulardii alone plus inulin resulted in the successful eradication of H. pylori in approximately 12% of children (Gotteland et al., 2005), and another study showed that L. reuteri combined with PPI therapy resulted in approximately 12.5% eradication of H. pylori (Dore et al., 2019). Although probiotics alone have some H. pylori eradication rates, they are not a substitute for the bactericidal role of antibiotics in eradication therapy, and supplementing probiotics to an eradication regimen has the potential to improve the effectiveness of antibiotic therapy. Table 3 summarizes the main results of four recently published meta-analysis studies on the effect of probiotic supplementation on H. pylori eradication, suggesting that probiotic supplementation as adjunctive therapy may improve eradication rates and reduce side effects in H. pylori eradication regimens (Shi et al., 2019; Yu et al., 2019; Zhou et al., 2019; Zhang et al., 2020). However, some studies have also shown that using probiotics in H. pylori eradication therapy is ineffective (Mcnicholl et al., 2018). This may be related to the probiotic supplement, the way and dosage used, the choice of antibiotics for eradication therapy, and the heterogeneity of the included patients. Moreover, diarrhea, sepsis, subacute bacterial endocarditis, and meningitis are all possible side effects of probiotics, but they are scarce (Doron and Snydman. Risk, 2015). Probiotics have potential risks that must be monitored. Thus, much more research is needed to determine the best probiotic and dose for specific diseases, first in animal models comparing different strains, and then in randomized controlled trials (Liu et al., 2018).

FIGURE 3
www.frontiersin.org

Figure 3 A schematic diagram of the potential mechanism of action of probiotics against H. pylori infection.

TABLE 3
www.frontiersin.org

Table 3 Main results of meta-analysis of the effect of probiotic supplementation on H. pylori eradication published in 2019-2020.

Scientific evidence of L. reuteri DSM 1764 for the relief of H. pylori infection

In 2002, a team led by Dr. Christine Lang, a pioneer in German microbiology research, screened more than 700 strains of Lactobacillus from its own library in vitro and identified a strain with a significant binding effect to H. pylori, L. reuteri DSM 17648 (Holz et al., 2015). A series of subsequent in vitro and in vivo studies showed that this strain specifically recognized and bound to the surface protein structure of H. pylori, thereby forming a co-polymer with H. pylori and excreting it through gastrointestinal motility, thereby reducing the H. pylori load in the stomach (Holz et al., 2015). In in vitro experiments, the aggregation of L. reuteri DSM 17648 with H. pylori strains can occur within seconds, and a single L. reuteri DSM 17648 cell can bind 2-3 H. pylori cells. Moreover, it binds specifically to several different H. pylori strains and other species of the genus Helicobacter, but not to other gastrointestinal pathogenic bacteria (e.g., C. jejuni) and oral and intestinal commensal bacteria (e.g., E. coli, S. salivarius, B. fragilis), suggesting that it should not disturb the normal intestinal flora balance. In addition, L. reuteri DSM 17648 effectively polymerizes H. pylori in an artificial gastric juice at 37°C. This action occurs in the pH range 2-8 (covering the pH range of gastric juice from fasting to postprandial) and is not interfered with by many common dietary sugar molecules, and requires the presence of pepsin to fully activate this polymerization activity. Unlike probiotics in the usual sense, L. reuteri DSM 17648 antagonizes H. pylori independently of the bacteria’s own activity - its aggregation activity against H. pylori remains effective after inactivation (e.g., spray drying treatment). This property not only reduces the difficulty of transport and storage but also suggests that L. reuteri DSM 17648 does not lose its ability to bind H. pylori when used in combination with antibiotics, increasing the feasibility of its use in combination with antibiotics.

Clinical trials of L. reuteri DSM 17648 for the treatment of H. pylori

The safety and efficacy of the strain have been validated in 12 clinical trials with 951 subjects across 6 countries: Germany, Ireland, Russia, Romania, China, and India (751 subjects involved in published trials so far and 200 subjects in progress) (Table 4).

TABLE 4
www.frontiersin.org

Table 4 Completed and ongoing clinical trials with 951 subjects in 6 countries: Germany, Ireland, Russia, Romania, China, and India on the safety and efficacy of L. reuteri DSM 17648 strain.

L. reuteri DSM 17648 alone reduces H. pylori infection load in adults

Three single-blind, placebo-controlled trials of adult patients infected with H. pylori published between 2013 and 2018, enrolling 73 patients, discovered that L. reuteri DSM 17648 alone significantly reduced H. pylori load in patients and helped reduce mild dyspeptic symptoms with a favorable safety profile (Mehling and Busjahn, 2013; Holz et al., 2015; Buckley et al., 2018). An uncontrolled intervention study of 60 infected adults published in 2016 also showed that L. reuteri DSM 17648 preparation reduced H. pylori load with a dose-dependent improvement (Bordin, 2015).

L. reuteri DSM 17648 for H. pylori eradication in adults, with long-term intervention success rates approaching those of triple therapy and substantially reduced treatment side effects

Two randomized controlled trials published in 2019 (Mihai, 2019; Muresan et al., 2019) enrolled 116 adult patients with H. pylori infection with functional dyspepsia and compared the efficacy of L. reuteri DSM 17648 formulation in combination with a PPI with conventional antibiotic therapy (triple therapy) for H. pylori eradication, antibiotic treatment side effects, and symptom improvement. L. reuteri DSM 17648 was found to be less effective than antibiotic therapy in eradicating H. pylori in the short term (14 days) and closer to antibiotic therapy in the long term (8 weeks) while having significantly lower side effects and improving symptoms. These results suggest that L. reuteri DSM 17648 may be a potential alternative treatment for H. pylori infection.

L. reuteri DSM 17648 improves gastrointestinal symptoms and reduces treatment side effects in infected adult with triple therapy

A prospective randomized controlled study reported in 2016 initially explored L. reuteri DSM 17648 in combination with triple therapy for treating H. pylori infection in 60 patients (Uspienskiy, 2016). This was followed by 2 randomized, double-blind controlled trials published in 2019 and 2021 (Parth et al., 2021; Yang et al., 2021), enrolling 290 adult patients with H. pylori infection. It was found that supplementing L. reuteri DSM 17648 to standard triple therapy may improve H. pylori eradication rates, improve patients’ gastrointestinal symptoms, reduce treatment-related side effects, and protect the intestinal flora. Notably, in both studies, Parth et al. showed that supplementation with L. reuteri DSM 17648 increased eradication rates by 20%. In contrast, Yang et al. showed no increase in eradication rates, a difference in results that may be related to differences in patient populations and treatment regimens. The higher eradication rate with triple therapy in Yang et al. may be related to population factors such as a lower mean age (mean age 30 years), which may have masked the effect of L. reuteri DSM 17648 to some extent. Further studies are needed to determine the effect of L. reuteri DSM 17648 supplementation on triple therapy eradication rates.

L. reuteri DSM 17648 reduces the load of H. pylori infection in children, improves symptoms, and reduces the side effects of treatment with quadruple therapy

Two clinical studies on L. reuteri DSM 17648 for the adjuvant treatment of children with chronic H. pylori infection, reported in 2015 (Ni, 2015) and 2020 (Kornienko et al., 2020), were conducted. A total of 152 children aged 9-17 years with H. pylori infection were enrolled in clinical studies of children with gastrointestinal diseases associated with H. pylori infection. The study found that long-term treatment (8 weeks) with L. reuteri DSM 17648 alone had an eradication rate comparable to that of quadruple therapy for long-term treatment (8 weeks) and improved gastrointestinal clinical symptoms and morphological changes in the gastric mucosa; when used in combination with quadruple therapy, it may increase eradication rates and reduce treatment side effects.

These clinical studies have shown that L. reuteri DSM 17648 formulation is safe and effective in reducing H. pylori load in adults and children with infection. When used alone, it has a lower eradication rate of H. pylori than conventional antibiotic therapy in the short term, but long-term use (e.g., for 8 weeks) may improve eradication rates to near that of antibiotic therapy; in addition, it has significantly fewer side effects and similar improvement in gastrointestinal symptoms compared to antibiotic therapy. When used in combination with antibiotic therapy, L. reuteri DSM 17648 has been shown to be effective in improving symptoms and reducing side effects and may further improve eradication rates. However, there are still some inconsistent results in the current studies due to differences in study design, subject populations, treatment regimens, and doses used in different trials. More high-quality, large-scale clinical trials need to be conducted in the future to validate.

Clinical trials have demonstrated that L. reuteri DSM 17648 alone can reduce the load of H. pylori infection and improve gastrointestinal symptoms in adults and children, while when used in combination with antibiotic therapy, L. reuteri DSM 17648 is effective in improving symptoms and adverse reactions in adults and children, and can further improve eradication rates and reduce treatment side effects. According to the findings of this study, L. reuteri DSM 17648 could be used as an adjunct to antibiotic therapy in the treatment of H. pylori infection and related diseases and may be an alternative therapy for those who are not candidates for antibiotic therapy. Since 2011, L. reuteri DSM 17648 and its use against H. pylori have applied for and received several patents for inventions granted in Europe, the United States, China, and Japan, as well as intellectual property rights for the trademark Pylopass™. In September 2016, Novozymes, a leading global provider of enzyme and microbial technologies, acquired ownership, patent rights, and a worldwide distribution license for L. reuteri DSM 17648 strain. This strain has been evaluated as a food ingredient safe for producing dietary supplements, health foods, and functional foods. Pylopass™ product is a spray-dried powder of L. reuteri DSM 17648 with dextrin as an excipient. By the end of 2021, there will be over 110 commercialized products containing Pylopass™ in numerous countries and regions worldwide. Of these, Europe is the most dominant origin with over 50%, and China is the second largest market with over 30 end-brand products.

Concluding remarks

As a global disease, diseases associated with H. pylori infection (gastritis, dyspepsia, peptic ulcer, gastric cancer, etc.) pose a heavy disease burden on human health. Over the last four decades, the prevalence of H. pylori infection has gradually declined as economic conditions and public health have improved. However, at least 40% of the population remains infected, with regional variations. Several mainstream international consensus guidelines recommend treating H. pylori infection with eradication. However, as H. pylori antibiotic resistance grows, the efficacy of conventional antibiotic eradication therapy declines. Recent studies suggest that specific probiotic strains may improve the efficacy of H. pylori eradication therapy. L. reuteri DSM 17648 (Pylopass™) is a probiotic strain that binds to H. pylori in the stomach and forms a copolymer. Several clinical trials have demonstrated that L. reuteri DSM 17648 is safe and effective in reducing H. pylori load and improving gastrointestinal symptoms in adults and children with the disease; when used in conjunction with antibiotic therapy, it may reduce adverse therapeutic effects and potentially improve eradication rates. However, the current clinical studies have limitations, such as small sample size and unblinded design. More high-quality, large-scale double-blind, randomized controlled trials should be conducted to validate the findings.

Probiotics directly compete with H. pylori to help restore the intestinal microbial environment and are more effective than standard triple therapy in treating H. pylori-related symptoms. The increasing rate of antibiotic resistance and decreased patient adherence to standard treatment better explain the need for alternative therapies. Adjunctive administration of probiotics to H. pylori eradication therapy was associated with higher H. pylori eradication rate, reduced diarrhea-related treatment, fewer common side effects, and higher treatment adherence. Thus, although the antagonist activity of probiotics is H. pylori strain-specific, with continued and future resistance to antibiotics, probiotics may become a future treatment trend when used alone or in combination with current guideline treatments such as adjuvant therapy, drug delivery systems, and boost of the immune system against H. pylori infection.

Author contributions

LB wrote the manuscript and made the figures and tables. D-MX contributed to the conception of the review. YY, P-XJ, L-XL, and H-XK contributed to the manuscript revision, adjusting figure images and improving the overall language. All authors listed have made a substantial, direct, and intellectual contribution to the work and approved it for publication.

Funding

This work is financially supported by the Qingdao Postdoctoral Applied Research Project (Grant No. RZ2200001423).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Abadi, A. T. B., Yamaoka, Y. (2018). Helicobacter pylori therapy and clinical perspective. J. Glob. Antimicrob. Re. 14, 111–117. doi: 10.1016/j.jgar.2018.03.005

CrossRef Full Text | Google Scholar

Agudo, S., Alarcon, T., Urruzuno, P., Martinez, M.J., Lopez-Brea, M. (2010). Detection of helicobacter pylori and clarithromycin resistance in gastric biopsies of pediatric patients by using a commercially available real-time polymerase chain reaction after NucliSens semiautomated DNA extraction. Diagn. Microbiol. Infect. Dis. 67, 213–219. doi: 10.1016/j.diagmicrobio.2010.02.021

PubMed Abstract | CrossRef Full Text | Google Scholar

Ansari, S., Yamaoka, Y. (2017). Survival of helicobacter pylori in gastric acidic territory. Helicobacter 22. doi: 10.1111/hel.12386

PubMed Abstract | CrossRef Full Text | Google Scholar

Bordin, D. S., Voynovan, I. N., Kolbasnikov, S.V. (2016). Evidence base of lactobacillus reuteri efficacy in the treatment of diseases associated with helicobacter pylori. Experimental Clin. Gastroenterol. (8), 82–87.

Google Scholar

Buckley, M., Lacey, S., Doolan, A., Goodbody, E., Seamans, K. (2018). The effect of lactobacillus reuteri supplementation in helicobacter pylori infection: a placebo-controlled, single-blind study. BMC Nutr. 4, 48. doi: 10.1186/s40795-018-0257-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, X., Wang, N., Wang, J., Liao, B., Cheng, L., Ren, B. (2022). The interactions between oral-gut axis microbiota and helicobacter pylori. Front. Cell Infect. Microbiol. 12, 914418. doi: 10.3389/fcimb.2022.914418

PubMed Abstract | CrossRef Full Text | Google Scholar

Chey, W. D., Leontiadis, G. I., Howden, C. W., Moss, S. F. (2017). ACG clinical guideline: Treatment of helicobacter pylori infection. Am. J. Gastroenterol. 112, 212–239. doi: 10.1038/ajg.2016.563

PubMed Abstract | CrossRef Full Text | Google Scholar

Chey, W. D., Wong, B. C., G Practice Parameters Committee of the American College Of (2007). American College of gastroenterology guideline on the management of helicobacter pylori infection. Am. J. Gastroenterol. 102, 1808–1825. doi: 10.1111/j.1572-0241.2007.01393.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Correa, P. (1992). Human gastric carcinogenesis: a multistep and multifactorial process–first American cancer society award lecture on cancer epidemiology and prevention. Cancer Res. 52, 6735–6740.

PubMed Abstract | Google Scholar

Datta, C., Subuddhi, A., Kumar, M., Lepcha, T.T., Chakraborty, S., Jana, K., et al. (2018). Genome-wide mRNA-miRNA profiling uncovers a role of the microRNA miR-29b-1-5p/PHLPP1 signalling pathway in helicobacter pylori-driven matrix metalloproteinase production in gastric epithelial cells. Cell Microbiol. 20, e12859. doi: 10.1111/cmi.12859

PubMed Abstract | CrossRef Full Text | Google Scholar

Dore, M. P., Bibbo, S., Pes, G. M., Francavilla, R., Graham, D.Y. (2019). Role of probiotics in helicobacter pylori eradication: Lessons from a study of lactobacillus reuteri strains DSM 17938 and ATCC PTA 6475 (Gastrus(R)) and a proton-pump inhibitor. Can. J. Infect. Dis. Med. Microbiol. 2019, 3409820. doi: 10.1155/2019/3409820

PubMed Abstract | CrossRef Full Text | Google Scholar

Dore, M. P., Lu, H., Graham, D. Y. (2016). Role of bismuth in improving helicobacter pylori eradication with triple therapy. Gut 65, 870–878. doi: 10.1136/gutjnl-2015-311019

PubMed Abstract | CrossRef Full Text | Google Scholar

Doron, S., Snydman. Risk, D. R. (2015). And safety of probiotics. Clin. Infect. Dis. 60 (Suppl 2), S129–S134. doi: 10.1093/cid/civ085

PubMed Abstract | CrossRef Full Text | Google Scholar

EaY Tshibangu-Kabamba, Y. (2021). Helicobacter pylori infection and antibiotic resistance - from biology to clinical implications. Nat. Rev. Gastroenterol. Hepatol. 18, 613–629. doi: 10.1038/s41575-021-00449-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Fakharian, F., Asgari, B., Nabavi-Rad, A., Sadeghi, A., Soleimani, N., Yadegar, A., et al. (2022). The interplay between helicobacter pylori and the gut microbiota: An emerging driver influencing the immune system homeostasis and gastric carcinogenesis. Front. Cell Infect. Microbiol. 12, 953718. doi: 10.3389/fcimb.2022.953718

PubMed Abstract | CrossRef Full Text | Google Scholar

Fallone, C. A., Chiba, N., Van Zanten, S. V., Fischbach, L., Gisbert, J. P., Hunt, R. H., et al. (2016). The Toronto consensus for the treatment of helicobacter pylori infection in adults. Gastroenterology 151, 51–69.e14. doi: 10.1053/j.gastro.2016.04.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Flores-Trevino, S., Mendoza-Olazaran, S., Bocanegra-Ibarias, P., Maldonado-Garza, H.J., Garza-Gonzalez, E.. (2018). Helicobacter pylori drug resistance: therapy changes and challenges. Expert Rev. Gastroent. 12, 819–827. doi: 10.1080/17474124.2018.1496017

CrossRef Full Text | Google Scholar

Fock, K. M., Katelaris, P., Sugano, K., Ang, T. L., Hunt, R., Talley, N. J., et al. (2009). Second Asia-pacific consensus guidelines for helicobacter pylori infection. J. Gastroenterol. Hepatol. 24, 1587–1600. doi: 10.1111/j.1440-1746.2009.05982.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Ford, A. C., Yuan, Y., Moayyedi, P. (2020). Helicobacter pylori eradication therapy to prevent gastric cancer: systematic review and meta-analysis. Gut 69, 2113–2121. doi: 10.1136/gutjnl-2020-320839

PubMed Abstract | CrossRef Full Text | Google Scholar

Forman, D., Webb, P., Parsonnet., J. (1994). H pylori and gastric cancer. Lancet 343, 243–244. doi: 10.1016/S0140-6736(94)91034-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Gonciarz, W., Walencka, M., Moran, A. P., Hinc, K., Obuchowski, M., Chmiela, M., et al. (2019). Upregulation of MUC5AC production and deposition of LEWIS determinants by HELICOBACTER PYLORI facilitate gastric tissue colonization and the maintenance of infection. J. BioMed. Sci. 26, 23. doi: 10.1186/s12929-019-0515-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Gotteland, M., Poliak, L., Cruchet, S., Brunser, O. (2005). Effect of regular ingestion of saccharomyces boulardii plus inulin or lactobacillus acidophilus LB in children colonized by helicobacter pylori. Acta Paediatr. 94, 1747–1751. doi: 10.1111/j.1651-2227.2005.tb01848.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Graham, D. Y., Lee, S. Y. (2015). How to effectively use bismuth quadruple therapy: The good, the bad, and the ugly. Gastroenterol. Clin. North Am. 44, 537–563. doi: 10.1016/j.gtc.2015.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, Y., Zhang, Y., Gerhard, M., Gao, J.J., Mejias-Luque, R., Zhang, L., et al. (2020). Effect of helicobacter pylori on gastrointestinal microbiota: a population-based study in linqu, a high-risk area of gastric cancer. Gut 69, 1598–1607. doi: 10.1136/gutjnl-2019-319696

PubMed Abstract | CrossRef Full Text | Google Scholar

Hamedi Asl, D., Naserpour Farivar, T., Rahmani, B., Hajmanoochehri, F., Emami Razavi, A.N., Jahanbin, B., et al. (2019). The role of transferrin receptor in the helicobacter pylori pathogenesis; l-ferritin as a novel marker for intestinal metaplasia. Microb. Pathog. 126, 157–164. doi: 10.1016/j.micpath.2018.10.039

PubMed Abstract | CrossRef Full Text | Google Scholar

Hathroubi, S., Zerebinski, J., Ottemann, K. M. (2018). Helicobacter pylori biofilm involves a multigene stress-biased response, including a structural role for flagella. mBio 9 (5), e01973-18. doi: 10.1128/mBio.01973-18

PubMed Abstract | CrossRef Full Text | Google Scholar

Hill, C., Guarner, F., Reid, G., Gibson, G.R., Merenstein, D.J., Pot, B., et al. (2014). Expert consensus document. the international scientific association for probiotics and prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 11, 506–514. doi: 10.1038/nrgastro.2014.66

PubMed Abstract | CrossRef Full Text | Google Scholar

Holz, C., Busjahn, A., Mehling, H., Arya, S., Boettner, M., Habibi, H., et al. (2015). Significant reduction in helicobacter pylori load in humans with non-viable lactobacillus reuteri DSM17648: A pilot study. Probiotics Antimicrob. Proteins 7, 91–100. doi: 10.1007/s12602-014-9181-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Homan, M., Orel, R. (2015). Are probiotics useful in helicobacter pylori eradication? World J. Gastroenterol. 21, 10644–10653. doi: 10.3748/wjg.v21.i37.10644

PubMed Abstract | CrossRef Full Text | Google Scholar

Hooi, J. K. Y., Lai, W. Y., Ng, W. K., Suen, M. M. Y., Underwood, F. E., Tanyingoh, D., et al. (2017). Global prevalence of helicobacter pylori infection: Systematic review and meta-analysis. Gastroenterology 153, 420–429. doi: 10.1053/j.gastro.2017.04.022

PubMed Abstract | CrossRef Full Text | Google Scholar

Ji, J., Yang, H. (2020). Using probiotics as supplementation for helicobacter pylori antibiotic therapy. Int. J. Mol. Sci. 21 (3), 1136. doi: 10.3390/ijms21031136

CrossRef Full Text | Google Scholar

Jones, M. D., Li, Y., Zamble, D. B. (2018). Acid-responsive activity of the helicobacter pylori metalloregulator NikR. Proc. Natl. Acad. Sci. U. S. A. 115, 8966–8971. doi: 10.1073/pnas.1808393115

PubMed Abstract | CrossRef Full Text | Google Scholar

Kienesberger, S., Cox, L. M., Livanos, A., Zhang, X.S., Chung, J., Perez-Perez, G. I., et al. (2016). Gastric helicobacter pylori infection affects local and distant microbial populations and host responses. Cell Rep. 14, 1395–1407. doi: 10.1016/j.celrep.2016.01.017

PubMed Abstract | CrossRef Full Text | Google Scholar

Kornienko, E. A., Ivanov, S. V. (2020). Gastric microbiota and probiotics opportunities in helicobacter pylori eradication in children. Gastroenterol Hepatol Open Access 11, 13–23. doi: 10.15406/ghoa.2020.11.00407

CrossRef Full Text | Google Scholar

Lind, T., Veldhuyzen Van Zanten, S., Unge, P., Spiller, R., Bayerdorffer, E., O'morain, C., 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. doi: 10.1111/j.1523-5378.1996.tb00027.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Liou, J. M., Malfertheiner, P., Lee, Y. C., Sheu, B.S., Sugano, K., Cheng, H.C., et al. (2020). Screening and eradication of helicobacter pylori for gastric cancer prevention: the Taipei global consensus. Gut 69, 2093–2112. doi: 10.1136/gutjnl-2020-322368

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, T. H., Qin, Y., Sham, P. C., Lau, K.S., Chu, K.M., Leung, W.K., et al. (2017). Alterations in gastric microbiota after h. pylori eradication and in different histological stages of gastric carcinogenesis. Sci. Rep. 7, 44935. doi: 10.1038/srep44935

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, L. P., Sheng, X. P., Shuai, T. K., Zhao, Y.X., Li, B., Li, Y.M. (2018). Helicobacter pylori promotes invasion and metastasis of gastric cancer by enhancing heparanase expression. World J. Gastroenterol. 24, 4565–4577. doi: 10.3748/wjg.v24.i40.4565

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Y., Tran, D. Q., Rhoads, J. M. (2018). Probiotics in disease prevention and treatment. J. Clin. Pharmacol. 58 Suppl 10, S164–SS79. doi: 10.1002/jcph.1121

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, W. Z., Xie, Y., Lu, H., Cheng, H., Zeng, Z.R., Zhou, L.Y., et al. (2018). Fifth Chinese national consensus report on the management of helicobacter pylori infection. Helicobacter 23, e12475. doi: 10.1111/hel.12475

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, L., Zhao, Y., Fan, G., Shuai, T., Li, B., Li, Y., et al. (2018). Helicobacter pylori infection enhances heparanase leading to cell proliferation via mitogenactivated protein kinase signalling in human gastric cancer cells. Mol. Med. Rep. 18, 5733–5741. doi: 10.3892/mmr.2018.9558

PubMed Abstract | CrossRef Full Text | Google Scholar

Llorca, L., Perez-Perez, G., Urruzuno, P., Martinez, M.J., Iizumi, T., Gao, Z., et al. (2017). Characterization of the gastric microbiota in a pediatric population according to helicobacter pylori status. Pediatr. Infect. Dis. J. 36, 173–178. doi: 10.1097/INF.0000000000001383

PubMed Abstract | CrossRef Full Text | Google Scholar

Malfertheiner, P., Bazzoli, F., Delchier, J. C., Celinski, K., Giguere, M., Riviere, 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. doi: 10.1016/S0140-6736(11)60020-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Malfertheiner, P., Megraud, F., O'morain, C., Bazzoli, F., El-Omar, E., Graham, D., et al. (2007). Current concepts in the management of helicobacter pylori infection: the maastricht III consensus report. Gut 56, 772–781. doi: 10.1136/gut.2006.101634

PubMed Abstract | CrossRef Full Text | Google Scholar

Malfertheiner, P., Megraud, F., O'morain, C. A., Atherton, J., Axon, A.T., Bazzoli, F., et al. (2012). Management of helicobacter pylori infection–the maastricht IV/ Florence consensus report. Gut 61, 646–664. doi: 10.1136/gutjnl-2012-302084

PubMed Abstract | CrossRef Full Text | Google Scholar

Malfertheiner, P., Megraud, F., O'morain, C. A., Gisbert, J.P., Kuipers, E.J., Axon, A.T., et al. (2017). Management of helicobacter pylori infection-the maastricht V/Florence consensus report. Gut 66, 6–30. doi: 10.1136/gutjnl-2016-312288

PubMed Abstract | CrossRef Full Text | Google Scholar

Matsunaga, S., Nishiumi, S., Tagawa, R., Yoshida, M. (2018). Alterations in metabolic pathways in gastric epithelial cells infected with helicobacter pylori. Microb. Pathog. 124, 122–129. doi: 10.1016/j.micpath.2018.08.033

PubMed Abstract | CrossRef Full Text | Google Scholar

Mcnicholl, A.G., Molina-Infante, J., Lucendo, A.J., Calleja, J.L., Perez-Aisa, A., Modolell, I., et al. (2018). Probiotic supplementation with lactobacillus plantarum and pediococcus acidilactici for helicobacter pylori therapy: A randomized, double-blind, placebo-controlled trial. Helicobacter 23, e12529. doi: 10.1111/hel.12529

PubMed Abstract | CrossRef Full Text | Google Scholar

Mehling, H., Busjahn, A. (2013). Non-viable lactobacillus reuteri DSMZ 17648 (Pylopass) as a new approach to helicobacter pylori control in humans. Nutrients 5, 3062–3073. doi: 10.3390/nu5083062

PubMed Abstract | CrossRef Full Text | Google Scholar

Mihai, C. (2019). Lactobacillus reuteri – an alternatie in the first-line in helicobacrer pylori eradication. FARMACIA 67 (5), 871–876. doi: 10.31925/farmacia.2019.5.17

CrossRef Full Text | Google Scholar

Mohsen, S., Dickinson, J. A., Somayaji, R. (2020). Update on the adverse effects of antimicrobial therapies in community practice. Can. Fam. Physician 66, 651–659. doi: 10.1111/hel.12386

PubMed Abstract | CrossRef Full Text | Google Scholar

Muresan, I., Pop, L. L., Dumitrascu, D. L. (2019). Lactobacillus reuteri versus triple therapy for the eradication of helicobacter pylori in functional dyspepsia. Med. Pharm. Rep. 92, 352–355. doi: 10.15386/mpr-1375

PubMed Abstract | CrossRef Full Text | Google Scholar

NCMRCFDD (Shanghai) (2021). Expert consensus on the prevention, control and management of helicobacter pylori infection in Chinese households (2021). Chin. J. Digest. 41, 221–233. doi: 10.3760/cma.j.cn311367-20210219-00108

CrossRef Full Text | Google Scholar

Nyssen, O. P., Perez-Aisa, A., Tepes, B., Castro-Fernandez, M., Kupcinskas, J., Jonaitis, L., et al. (2021). Adverse event profile during the treatment of helicobacter pylori: A real-world experience of 22,000 patients from the European registry on h. pylori management (Hp-EuReg). Am. J. Gastroenterol. 116, 1220–1229. doi: 10.14309/ajg.0000000000001246

PubMed Abstract | CrossRef Full Text | Google Scholar

O'connor, A., O'morain, C. A., Ford, A. C. (2017). Population screening and treatment of helicobacter pylori infection. Nat. Rev. Gastroenterol. Hepatol. 14, 230–240. doi: 10.1038/nrgastro.2016.195

PubMed Abstract | CrossRef Full Text | Google Scholar

Parolova, N. I., Kornienko, E. A., Antonov, P. V., Egorov, M., Gorbunov, E. F., Dmitrienko, M. A., et al. (2015). An innovative approach in the treatment of H. pylori infection in children. RMJ 22, 1339–1340.

Google Scholar

Parth, K., Prudhivi, R., Palatheeya, S., Abbas, S. K., Varsha, K., Niharika, B., et al. (2021). Efficacy of lactobacillus reuteri supplementation in eradication of h. pylori: A comparison study with triple drug therapy. J. Pharm. Res. Int. doi: 10.9734/jpri/2021/v33i52b33611

PubMed Abstract | CrossRef Full Text | Google Scholar

Qureshi, N., Li, P., Gu, Q. (2019). Probiotic therapy in helicobacter pylori infection: a potential strategy against a serious pathogen? Appl. Microbiol. Biotechnol. 103, 1573–1588. doi: 10.1007/s00253-018-09580-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Ronci, M., Del Prete, S., Puca, V., Carradori, S., Carginale, V., Muraro, R., et al. (2019). Identification and characterization of the alpha-CA in the outer membrane vesicles produced by helicobacter pylori. J. Enzyme Inhib. Med. Chem. 34, 189–195. doi: 10.1080/14756366.2018.1539716

PubMed Abstract | CrossRef Full Text | Google Scholar

Saenz, J. B., Vargas, N., Mills, J. C. (2019). Tropism for spasmolytic polypeptide-expressing metaplasia allows helicobacter pylori to expand its intragastric niche. Gastroenterology 156, 160–74 e7. doi: 10.1053/j.gastro.2018.09.050

PubMed Abstract | CrossRef Full Text | Google Scholar

Savoldi, A., Carrara, E., Graham, D. Y., Conti, M., Tacconelli, E. (2018). Prevalence of antibiotic resistance in helicobacter pylori: A systematic review and meta-analysis in world health organization regions. Gastroenterology 155, 1372–82.e17. doi: 10.1053/j.gastro.2018.07.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Schmidt, T. S. B., Raes, J., Bork, P. (2018). The human gut microbiome: From association to modulation. Cell 172, 1198–1215. doi: 10.1016/j.cell.2018.02.044

PubMed Abstract | CrossRef Full Text | Google Scholar

Shi, X., Zhang, J., Mo, L., Shi, J., Qin, M., Huang, X. (2019). Efficacy and safety of probiotics in eradicating helicobacter pylori: A network meta-analysis. Med. (Baltimore) 98, e15180. doi: 10.1097/MD.0000000000015180

CrossRef Full Text | Google Scholar

Smyth, E. C., Nilsson, M., Grabsch, H. I., Van Grieken, N. C., Lordick, F. (2020). Gastric cancer. Lancet 396, 635–648. doi: 10.1016/S0140-6736(20)31288-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Sticlaru, L., Staniceanu, F., Cioplea, M., Nichita, L., Bastian, A., Micu, G., et al. (2018). Neuroimmune crosstalk in helicobacter pylori infection: analysis of substance p and vasoactive intestinal peptide expression in gastric enteric nervous system. J. Immunoassay Immunochem. 39, 660–671. doi: 10.1080/15321819.2018.1529683

PubMed Abstract | CrossRef Full Text | Google Scholar

Suerbaum, S., Michetti, P. (2002). Helicobacter pylori infection. N Engl. J. Med. 347, 1175–1186. doi: 10.1056/NEJMra020542

PubMed Abstract | CrossRef Full Text | Google Scholar

Suez, J., Zmora, N., Segal, E., Elinav, E. (2019). The pros, cons, and many unknowns of probiotics. Nat. Med. 25, 716–729. doi: 10.1038/s41591-019-0439-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Sugano, K., Tack, J., Kuipers, E. J., Graham, D.Y., El-Omar, E.M., Miura, S., et al. (2015). Kyoto Global consensus report on helicobacter pylori gastritis. Gut 64, 1353–1367. doi: 10.1136/gutjnl-2015-309252

PubMed Abstract | CrossRef Full Text | Google Scholar

Tacconelli, E., Carrara, E., Savoldi, A., Harbarth, S., Mendelson, M., Monnet, D. L., et al. (2018). Discovery, research, and development of new antibiotics: the WHO priority list of antibiotic-resistant bacteria and tuberculosis. Lancet Infect. Dis. 18, 318–327. doi: 10.1016/S1473-3099(17)30753-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Uspienskiy, Y. P., Fomin, Y. A., Ivanov, S.V., Menaker, I.O. (2016). Evolution in eradication therapy of hp-associated diseases: beyond the standards? RMJ 17, 3–1152.

Google Scholar

Wen, G., Deng, S., Song, W., Jin, H., Xu, J., Liu, X., et al. (2018). Helicobacter pylori infection downregulates duodenal CFTR and SLC26A6 expressions through TGFbeta signaling pathway. BMC Microbiol. 18, 87. doi: 10.1186/s12866-018-1230-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, C., Liang, L., Lv, P., Liu, L., Wang, S., Wang, Z., et al. (2021). Effects of non-viable lactobacillus reuteri combining with 14-day standard triple therapy on helicobacter pylori eradication: A randomized double-blind placebo-controlled trial. Helicobacter 26, e12856. doi: 10.1111/hel.12856

PubMed Abstract | CrossRef Full Text | Google Scholar

Yao X.J, L. Y. (2018). Effect of modified sanhuang xiexin tang plus additional herbs combined with “standard triple therapy” on helicobacter pylori eradication. J. Tradit. Chin. Med. 38, 101–106. doi: 10.1016/j.jtcm.2018.02.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Yaseen, A., Audette, G. F. (2018). Structural flexibility in the helicobacter pylori peptidyl-prolyl cis,trans-isomerase HP0175 is achieved through an extension of the chaperone helices. J. Struct. Biol. 204, 261–269. doi: 10.1016/j.jsb.2018.08.017

PubMed Abstract | CrossRef Full Text | Google Scholar

Yokota, S., Konno, M., Fujiwara, S., Toita, N., Takahashi, M., Yamamoto, S., et al. (2015). Intrafamilial, preferentially mother-to-Child and intraspousal, helicobacter pylori infection in Japan determined by mutilocus sequence typing and random amplified polymorphic DNA fingerprinting. Helicobacter 20, 334–342. doi: 10.1111/hel.12217

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, M., Zhang, R., Ni, P., Chen, S., Duan, G. (2019). Efficacy of lactobacillus-supplemented triple therapy for h. pylori eradication: A meta-analysis of randomized controlled trials. PloS One 14, e0223309. doi: 10.1371/journal.pone.0223309

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, W., Chen, Q., Liang, X., Liu, W., Xiao, S., Graham, D. Y., et al. (2015). Bismuth, lansoprazole, amoxicillin and metronidazole or clarithromycin as first-line helicobacter pylori therapy. Gut 64, 1715–1720. doi: 10.1136/gutjnl-2015-309900

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, S., Shi, D., Li, M., Li, Y., Wang, X., Li, W. (2019). The relationship between gastric microbiota and gastric disease. Scand. J. Gastroenterol. 54, 391–396. doi: 10.1080/00365521.2019.1591499

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, X. Z. W. W., Lan, Y. (2020). Expert consensus on joint diagnosis and treatment of gastritis caused by helicobacter pylori in adults (2020, Beijing). J. Tradit. Chin. Med. 61 (22), 2016–2024. doi: 10.13288/j.11-2166/r.2020.22.019

CrossRef Full Text | Google Scholar

Zhang, M., Zhang, C., Zhao, J., Zhang, H., Zhai, Q., Chen, W.. (2020). Meta-analysis of the efficacy of probiotic-supplemented therapy on the eradication of h. pylori and incidence of therapy-associated side effects. Microb. Pathog. 147, 104403. doi: 10.1016/j.micpath.2020.104403

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, B. G., Chen, L. X., Li, B., Wan, L. Y., Ai, Y. W. (2019). Saccharomyces boulardii as an adjuvant therapy for helicobacter pylori eradication: A systematic review and meta-analysis with trial sequential analysis. Helicobacter 24, e12651. doi: 10.1111/hel.12651

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: Helicobacter pylori, antibiotic resistance, eradication therapy, probiotics, Limosilactobacillus reuteri DSM 17648

Citation: Liang B, Yuan Y, Peng X-J, Liu X-L, Hu X-K and Xing D-M (2022) Current and future perspectives for Helicobacter pylori treatment and management: From antibiotics to probiotics. Front. Cell. Infect. Microbiol. 12:1042070. doi: 10.3389/fcimb.2022.1042070

Received: 12 September 2022; Accepted: 02 November 2022;
Published: 25 November 2022.

Edited by:

Rosa Sessa, Sapienza University of Rome, Italy

Reviewed by:

Xuesong Sun, Jinan University, China
Amin Talebi Bezmin Abadi, Tarbiat Modares University¸, Iran

Copyright © 2022 Liang, Yuan, Peng, Liu, Hu and Xing. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Dong-Ming Xing, xdm_tsinghua@163.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.