Journal List > J Korean Med Sci > v.39(4) > 1516086185

Hasanuzzaman, Bang, and Gong: Antibiotic Resistance of Helicobacter pylori: Mechanisms and Clinical Implications

Abstract

Helicobacter pylori is a pathogenic bacterium associated with various gastrointestinal diseases, including chronic gastritis, peptic ulcers, mucosa-associated lymphoid tissue lymphoma, and gastric cancer. The increasing rates of H. pylori antibiotic resistance and the emergence of multidrug-resistant strains pose significant challenges to its treatment. This comprehensive review explores the mechanisms underlying the resistance of H. pylori to commonly used antibiotics and the clinical implications of antibiotic resistance. Additionally, potential strategies for overcoming antibiotic resistance are discussed. These approaches aim to improve the treatment outcomes of H. pylori infections while minimizing the development of antibiotic resistance. The continuous evolution of treatment perspectives and ongoing research in this field are crucial for effectively combating this challenging infection.

INTRODUCTION

Helicobacter pylori is a Gram-negative bacterium that colonizes and persists in the stomach.1 It can be transmitted from an infected person to an uninfected person by direct contact via an oral-oral, fecal-oral, or both routes.23 Most people become infected during their childhood, and parents and siblings appear to play a significant role in pathogen transmission.34 Once infected, H. pylori causes lifelong chronic progressive gastric inflammation, which can lead to clinical complications in up to 10% of infected individuals.15 The major clinical complications include peptic ulcer disease, chronic gastritis, gastric cancer, and mucosa-associated lymphoid tissue lymphoma.12567 The prevalence of H. pylori infection varies widely according to geographic area, age, and socioeconomic status.6 Although the prevalence of H. pylori is decreasing due to improving hygiene and standard of living, it remains high, particularly in the Eastern Asian countries.68910
A combination of two to three antibiotics (from a few antibiotics such as amoxicillin, clarithromycin, metronidazole, tetracycline, levofloxacin, and rifabutin) and an acid-suppressive agent with or without bismuth are used to eradicate H. pylori.111213 However, the successful eradication rate of H. pylori has decreased in the past decades, in parallel with increasing antibiotic resistance.14151617181920 Antibiotic resistance of H. pylori is particularly important because this is one of the most common causes of bacterial infections worldwide, affecting millions of people every year.16 Additionally, overuse or inappropriate use of antibiotics can contribute to the development of antimicrobial resistance in H. pylori and other bacteria, which can have serious public health implications.212223 Recently, the World Health Organization listed H. pylori as a serious threat to human health for their resistance against most available treatment regimens.24
The molecular mechanisms underlying antibiotic resistance in H. pylori infection are diverse and complex. Several mechanisms have been proposed to drive the antibiotic resistance of H. pylori, including genetic mutations in the bacterium itself and physiological changes that can upregulate efflux pump expression in bacterial cells.252627282930 Cellular adaptation associated with biofilm or coccoid formation, which protects drug penetration into bacterial cells, is another potential resistance mechanism.3132 Many of these resistance mechanisms can work in concert to confer multidrug resistance (MDR) in H. pylori, making eradication increasingly challenging and highlighting the need for new therapeutic strategies. In this review, the mechanism of resistance of H. pylori to commonly used antibiotics and their clinical implications are explored.

MOLECULAR MECHANISMS OF ANTIBIOTIC RESISTANCE

Mechanism of single-drug resistance

Single-drug resistance refers to a situation in which a microorganism, such as a bacterium, virus, or fungus, is resistant to only one type of drug while remaining susceptible to other drugs. Although the eradication regimen for H. pylori consists of a combination of antibiotics, resistance to a single antibiotic agent could result in eradication failure.33 Antibiotic resistance in H. pylori is mainly from de novo genetic mutations that disrupt the activity of antibiotics by either altering the drug target or inhibiting drug activation within cells (Table 1 and Fig. 1).7 With advances in next-generation sequencing (NGS), many mutations have been observed in resistant clinical isolates.25263435 However, the relative contribution of each mutation to phenotypic resistance and the effect of combinations of mutations remain unclear.36
Table 1

Mechanisms of antibiotic resistance in H. pylori

jkms-39-e44-i001
Antibiotics Antibiotic resistance mechanism Associated gene or sequence
Amoxicillin Mutational change of penicillin-binding protein pbp1A, pbp2, pbp3, pbp4
Production of beta-lactamase
Outer membrane protein mutations causing decreased membrane permeability hefC, hopC, hofH
Clarithromycin Point mutations in 23S rRNA 23S rRNA
Increased antibiotic efflux mediated by efflux pump systems rpl22, infB
Levofloxacin Point mutations in quinolone resistance determination region of DNA gyrase gene gyrA, gyrB
Metronidazole Mutations causing reduced or abolished nitroreductase activity rdxA, frxA
Tetracycline Mutations in 16S rRNA 16S rRNA
Rifabutin Point mutations in rifampicin-resistance determining region of RNA polymerase gene rpoB
Fig. 1

Overview of molecular mechanism of antibiotic resistance in H. pylori and strategies to overcome this resistance. This image was created with BioRender.com.

FrxA = NAD(P)H flavin oxidoreductase, GI = gastrointestinal, GyrA = DNA gyrase subunit A, GyrB = DNA gyrase subunit B, PBP = penicillin-binding protein, PCAB = potassium-competitive acid blocker, RdxA = oxygen-insensitive NAD(P)H nitroreductase, RpoB = β-subunit of DNA-dependent RNA polymerase, rRNA = ribosomal RNA, tRNA = transfer RNA.
jkms-39-e44-g001

Beta-lactams

Amoxicillin is widely used for H. pylori eradication in combination with acid-suppressive agents to improve drug stability and efficacy. After administration, amoxicillin is well absorbed into bloodstream and then released into the gastric juice.37 In a favorable environment with acid-suppressive agents, amoxicillin exerts its antimicrobial effect by binding to penicillin-binding proteins (PBPs).3839 Binding of amoxicillin to PBPs inhibits the synthesis of peptidoglycan, a major component of bacterial cell walls, resulting in cell wall lysis in replicating bacteria.3940 Amoxicillin generally shows a low antibiotic resistance rate.41424344 In H. pylori, amoxicillin resistance is mainly due to mutational changes in PBP1A.45464748 Mutations in other PBPs (PBP2 and PBP 3) have also reported, and multiple mutations in all three isotypes PBP1, PBP2 and PBP3 was found to confer high level of amoxicillin resistance when compared to the amoxicillin susceptible strain.49 Beta-lactamase activity, which can hydrolyze amoxicillin to attenuate the optimum concentration needed to elicit bactericidal effect, was also found in H. pylori.4050 Furthermore, amoxicillin resistance may be contributed by mutations in hefC, hopC, and hofH, which are likely associated with changes in the composition of the outer membrane and membrane permeability of H. pylori.515253

Macrolides

Clarithromycin is widely used in the frontline regimen to eradicate H. pylori. Clarithromycin has pharmacokinetic advantages over other macrolides, including increased oral bioavailability, higher plasma concentration, and longer elimination half-life.54 Concomitant administration with acid-suppressive agents also increases its stability in acidic environments.55 Clarithromycin exerts antimicrobial effects by binding to the peptidyl transferase loop of domain V in the 23S ribosomal RNA (rRNA) in the bacterial ribosomal subunit 50S. Mutations in domain V of the 23S rRNA gene of H. pylori, A2142G/C and A2143G, can result in reduced binding affinity of the antibiotic agent, making it less effective at inhibiting bacterial growth.5657 Other point mutations are also reported in the 23S rRNA in H. pylori isolates.25262758 In addition, studies using experimentally induced resistant phenotype for clarithromycin found that mutations in rpl22 and infB genes had synergistic effects with mutations in the 23S rRNA genes, resulting in higher minimum inhibitory concentration of clarithromycin.3435 Another relevant mechanism for clarithromycin resistance is attributed to the efflux pump system.2959 However, the role of novel mutations and specific function of efflux pump system in the development of clarithromycin resistance in clinical isolates should be further clarified.

Fluoroquinolones

Among the fluoroquinolones, levofloxacin, moxifloxacin, and sitafloxacin have been used for H. pylori eradication therapy. Due to high resistance rate, fluoroquinolones are generally used in rescue treatment after initial eradication failure.116061 Fluoroquinolones act on microbes by inhibiting bacterial topoisomerase II (DNA gyrase) and topoisomerase IV enzymes involved in bacterial synthesis of nucleic acid, a step proceeding cell division and proliferation.28 The most common mechanism of fluoroquinolone resistance in H. pylori is due to a specific mutation in one or more of gyrA and gyrB genes.62 The region where mutations arise in these genes is a short DNA sequence known as the quinolone resistance-determining region (QRDR).6364 Fluoroquinolone resistance of H. pylori is mainly due to point mutations of codon position 87 and 91 in the QRDR of gyrA.656667 These few mutations associated with most cases of phenotypic resistance indicate that molecular testing for levofloxacin can be a reliable substitute for culture and antimicrobial susceptibility testing. Mutations present outside the QRDR region of gyrA or in the QRDR region of gyrB have also been reported to be associated with levofloxacin resistance; however, the impact of these mutations requires further investigation.26275866

Nitroimidazole

Among the nitroimidazoles, metronidazole is frequently used to eradicate H. pylori infections. Metronidazole is actively released into gastric juice after oral ingestion, with the acidic condition in the stomach rarely affecting its antimicrobial activity.68 Metronidazole is a prodrug that needs to be activated by intracellular reduction of the nitro group attached to the imidazole ring.69 Reductive activation of metronidazole causes imidazole fragmentation and nitro-anion free radicals which are cytotoxic.6970 The reduction of metronidazole is mainly mediated by oxygen-insensitive NAD(P)H nitroreductase (RdxA), NAD(P)H flavin oxidoreductase (FrxA), and ferredoxin-like enzymes (FdxB) in H. pylori. Metronidazole resistance in H. pylori is primarily due to decreased drug activation mediated by mutations in RdxA gene which encodes an oxygen-insensitive NAD(P)H nitroreductase.70717273747576 Mutations involving the FrxA gene were also reported.357477 However, metronidazole resistance was observed in H. pylori isolates without the loss of functional RdxA and FrxA, suggesting that other factors are involved in metronidazole resistance.7278798081 Other putative mechanisms of metronidazole resistance in H. pylori include mutations in FdxB, ferric uptake regulator (Fur), and enhancement of efflux pump (HefA) protein.72828384

Tetracyclines

Tetracycline is stable in gastric pH and acts as a topical agent on the surface of the gastric mucosa against H. pylori.85 At the bacterial cytoplasm, tetracycline binds to bacterial ribosomes and interacts with a highly conserved 16S rRNA target in the 30S ribosomal subunit, arresting translation and protein synthesis.86 Resistance mechanism of H. pylori against tetracycline is not widely studied because tetracycline resistance is not common in clinical isolates.14414487 Among various mechanisms of tetracycline resistance, the major resistance mechanisms are related to mutations in the 16S rRNA genes.888990 However, tetracycline resistance without mutation in the 16S rRNA gene has been reported, suggesting that other mechanisms, such as efflux, are associated with tetracycline resistance.8491

Rifamycins

Rifamycins are transcriptional inhibitors that specifically inhibit the activity of bacterial transcription by binding to RNA polymerase, mostly β-subunit encoded by the rpoB gene.92 Among the rifamycins, rifabutin has better pharmacokinetics than rifampicin and is used for H. pylori eradication. Rifabutin is chemically stable at a wide range of pH values and is not inactivated by gastric acid.93 The resistance rate of H. pylori against rifabutin is low, and most rifabutin-resistant strains were isolated after treatment failure.949596 In H. pylori, the molecular mechanism driving rifabutin resistance is at least one point mutation in the rifampicin resistance-determining region of the rpoB gene.27979899100 However, a previous study reported that rifabutin-resistant strains were successfully eradicated with rifabutin-based triple therapy.101 Correlations between rpoB gene mutation status, phenotypic resistance, and treatment outcome need further clarification.

MDR in H. pylori

The presence of H. pylori strains with MDR profiles poses a significant challenge for H. pylori eradication and complicates the management of H. pylori-related diseases. The MDR profile of H. pylori is expressed as the cumulative result of genetic mutations conferring resistance to each antibiotic agent.7 Other putative mechanisms of H. pylori MDR include physiologic changes in bacterial cells (upregulation of efflux pump systems or downregulation of drug uptake proteins in the outer membrane of bacteria) and cellular adaptation properties (biofilm or coccoid formation).59102103104105
Bacterial biofilms are complex microbiological ecosystems where adherent aggregates of microorganisms surround themselves in multidimensional extracellular polymeric substances.106107 Biofilms are often associated with chronic infectious diseases as they protect the bacteria from unfavorable environments, antimicrobial exposure, and host immune system.31108 Biofilm formation in H. pylori has been observed both in vitro (environmental water body) and in vivo (gastric mucosa).109110 The presence of biofilm was shown to be associated with decreased susceptibility to antibiotics in H. pylori.31 Previous studies showed that mutations in several genes coding for flagellar protein, outer membrane protein, cytotoxin-associated gene pathogenicity island protein, or efflux pumps were responsible for biofilm formation in H. pylori, which can further potentiate antibiotic resistance.84111112 However, the precise mechanisms for H. pylori biofilm formation have yet to be determined. In addition, the clinical implications of biofilm formation on the development of antibiotic resistance in H. pylori and treatment outcomes should be further investigated.
An exceptional feature of H. pylori is the formation of a viable but non-culturable coccoid morphology. In H. pylori, the coccoid form becomes dominant when bacteria are exposed to environmental stress conditions, such as starvation, prolonged culture, and exposure to antibiotics.113114 It has been reported that this dormant state of H. pylori induces ultrastructural modifications in the cell membrane and metabolic pathways that contribute to antibiotic resistance.24107 However, the clinical relevance of coccoid formation in the development of MDR profile and treatment outcome of H. pylori is not fully understood.

Heteroresistance

Heteroresistance refers to a phenomenon where subpopulations of bacteria have different antibiotic susceptibility profiles.115 Heteroresistance in H. pylori has been reported in several studies in which both susceptible and resistant bacterial strains were isolated either from the same biopsy site (intraniche) or from different sites (interniche).87115116117118119120121122 Heteroresistance can be developed through evolutionary change in a single strain or mixed infection of multiple bacterial strains.7 Previous studies reported that heteroresistant H. pylori strains had similar fingerprinting patterns, suggesting that the presence of the same strain with mixed susceptible and resistant phenotype, rather than coinfection of different strains, is associated with the development of H. pylori heteroresistance.117118119120121 The possibility of heteroresistance should be considered during antimicrobial susceptibility testing and eradication therapy for H. pylori infection since underestimation of the presence of antibiotic-resistant strain may lead to treatment failure. To address this issue, multiple biopsies from different sites in the stomach or multiple bacterial colonies from the same sample should be obtained when evaluating the antimicrobial susceptibility of H. pylori.

Antimicrobial susceptibility testing

With increasing antibiotic resistance, it is imperative to develop individualized therapeutic approaches based on the results of antimicrobial susceptibility tests. Common methods for antimicrobial susceptibility testing include culture-based antimicrobial susceptibility testing and molecular detection. Bacterial culture is necessary for the use of conventional antimicrobial susceptibility tests, such as E-test, disk diffusion method, and agar dilution method.3942123 However, H. pylori culture requires specific conditions, is time-consuming, and is affected by several factors, such as transport conditions and the time interval between specimen collection and inoculation, limiting its availability in clinical practice.
Molecular methods are also used to assess antimicrobial susceptibility. Since the resistance of H. pylori to clarithromycin, fluoroquinolones, and tetracycline is mainly driven by specific point mutations in a small region of the responsible gene, molecular methods can be utilized for antimicrobial susceptibility testing. Polymerase chain reaction (PCR) was used to assess antibiotic resistance by detecting resistance-associated mutations. Similarly, PCR-restriction fragment length polymorphism, real-time PCR, multiplex PCR, and droplet digital PCR have been widely used to determine antibiotic resistance in H. pylori.124125126127128129 Currently, several commercial kits are available to detect clarithromycin resistance, levofloxacin resistance, or both.130131132133134135 Another diagnostic tool using loop-mediated isothermal amplification methods, combined PCR and quenching probe method, and single cell-based antimicrobial susceptibility test Ramanometry has been introduced.136137138139
Compared to conventional methods, molecular detection methods have the advantages of high sensitivity, specificity, and reproducibility, and disadvantages such as high cost. Furthermore, as molecular detection methods target specific gene loci, antibiotic resistance caused by other mutations cannot be detected, leading to false-negative results.140 In addition, as metronidazole has complex resistance mechanisms and the presence of metronidazole resistance does not necessarily lead to treatment failure, the role of molecular detection of metronidazole resistance is limited.
Recent advances in high-throughput molecular detection technology, including NGS and metagenomic analysis, have enabled the detection of H. pylori infection and antibiotic resistance.25262734 While NGS provides comprehensive information, applying NGS results to clinical practice requires additional knowledge about the correlation between NGS results and phenotypic resistance as well as treatment outcomes.141 In addition, cost-effectiveness and availability should be considered for the clinical application of NGS to predict antibiotic susceptibility profiles.

CILNICAL IMPLICATION AND FUTURE DIRECTIONS

The main clinical implication of antibiotic resistance in H. pylori is significantly compromised eradication therapy efficacy. It is well-known that the eradication success rate using clarithromycin-based regimens is markedly decreased in the presence of clarithromycin resistance.33142143 Accordingly, clarithromycin containing standard triple therapy is recommended only in areas with a clarithromycin resistance rate of less than 15%.11 On the other hand, a lower decrease in eradication rates has been observed for metronidazole, and successful eradication was reported using bismuth quadruple therapy, especially with higher metronidazole dose, even in the presence of metronidazole resistance.33142143 Multiple different mechanisms leading to metronidazole resistance and diversity of identified mutations may explain the relatively poor correlation between phenotypic resistance against metronidazole and treatment outcome. The increasing number of MDR H. pylori strains has made eradication therapy more challenging. Given that the first-line treatment regimen is usually selected empirically rather than based on antimicrobial susceptibility testing, failure of initial eradication therapy is another important cause of the emergence of MDR H. pylori strains.
The development of antibiotic resistance in H. pylori has reduced available treatment options. Eradication failure necessitates additional rounds of therapy, including alternative antibiotic combinations. With the decline in the eradication rates of standard therapies, physicians face challenges in selecting effective alternatives. However, these options are not always effective, involve higher pill burdens, and can lead to more side effects. There have been some important changes to expert recommendations for H. pylori eradication: more frequent use of bismuth-containing quadruple therapy as a first-line treatment instead of clarithromycin-based triple therapy is recommended.144 The limited arsenal of effective treatments against antibiotic-resistant H. pylori strains underscores the need for novel treatment strategies, including non-antibiotic approaches, to address this growing problem (Fig. 1).

Optimizing eradication therapy

Current treatment regimens for H. pylori eradication are derived from empirical approaches developed by gastroenterologists over the past few decades. A significant decrease in the eradication rate worldwide, along with an increasing trend of H. pylori antibiotic resistances warrants more specific approach to H. pylori eradication therapy, utilizing antimicrobial stewardship.145 Given that antibiotic resistance patterns differ according to geographic regions, it is recommended to use treatment regimens that are locally effective according to regional susceptibility patterns.87145146 However, many regions lack reliable data on the prevalence and characteristics of antibiotic resistance in local populations to guide the selection of empirical eradication therapy. Currently, bismuth quadruple therapy is recommended in regions with high antibiotic resistance to both clarithromycin and metronidazole.11 In addition, adding bismuth to some triple regimens and prolonging the treatment duration to 14 days can increase eradication rates up to 30% or more, even in the presence of antibiotic resistance in some strains.13 Recent meta-analysis also showed that bismuth supplements as the first-line regimen showed better eradication rate compared to non-bismuth containing regimens.147

Tailored therapy

With increasing resistance rates and MDR H. pylori strains, the role of antimicrobial susceptibility testing and subsequent individualized antibiotic treatment has been emphasized.11 Tailored therapy, in which antibiotics are chosen based on the antimicrobial susceptibility profile, is an ideal therapeutic option to improve the efficacy of H. pylori eradication therapy while minimizing unnecessary prescription of antibiotics. Studies have reported better eradication success rates using tailored therapy than empiric therapy, especially when antimicrobial susceptibility testing was performed before treatment.148149150151 However, the benefit of tailored therapy has not been demonstrated in clinical trials comparing tailored therapy with empirical quadruple regimens as a first-line treatment or tailored therapy after previous treatment failure.151152153 This suggests that the benefit of tailored therapy is not obvious when the empirical regimen is highly effective for H. pylori eradication. In addition, the limited availability of H. pylori cultures and antimicrobial susceptibility testing has made this approach difficult. More evidence is required to establish the generalized use of tailored therapy for H. pylori eradication.

Acid suppression with potassium-competitive acid blocker

To increase eradication success, antibiotic resistance patterns and patient characteristics, such as compliance and cytochrome P450 2C19 genetic polymorphisms, should be considered. Potassium-competitive acid blocker provides fast and long-lasting acid suppression and is recently used for H. pylori eradication therapy in combination with various antibiotics.154155156157 Vonoprazan-based triple therapy showed higher eradication rate compared to proton pump inhibitor (PPI), especially in the subpopulation with clarithromycin resistance, potentially overcoming clarithromycin resistance.158159 Recent meta-analysis of randomized controlled trials also showed that vonoprazan-containing regimens achieved a higher eradication success rate compared to PPI-based regimens.160 In line with this, a Japanese guideline recommends vonoprazan-based triple therapy or PPI-based triple therapy as the first-line treatment for H. pylori eradication.161 Vonoprazan-based dual therapy consisting amoxicillin may be another treatment option while minimizing unnecessary antibiotic use; however, the dosage and duration of dual therapy need to be further determined.159162163 Notably, most studies on vonoprazan-based regimens were conducted in Japan, and further studies are needed to confirm these promising results in different regions.164165

Adjuvant therapies

Adjuvant therapies aim to enhance the efficacy of antibiotic treatment either by overcoming the bacterial mechanisms of antibiotic resistance or by modifying the host response. These include the use of probiotics and anti-biofilm agents. As supplementary to conventional eradication therapy, probiotics can reduce gastrointestinal adverse events and thus improve patient compliance.166167 In a recent meta-analysis, it has been found that most probiotics added to triple therapy provided better treatment outcomes.167 The potential mechanisms of probiotic action against H. pylori include direct and indirect inhibitory effects against H. pylori colonization, stabilization of gastric mucosal protective barrier and reduction of gastric mucosal inflammation, and modulation of host immune response to the infection.168169 Probiotic supplementation also reduced antibiotic-induced alteration of gut microbiota and helped the restoration of dysbiosis caused by eradication therapy.170171 However, further research is needed to better understand the role and mechanism of action of probiotics in H. pylori eradication therapy, as the species and strains, dose, and duration of investigated probiotics are heterogeneous.
An alternative therapeutic approach is to target and disrupt bacterial biofilms. Previous studies have shown that the use of N-acetylcysteine (NAC) reduces bacterial load and enhances eradication rate.172173 The effect of NAC was also found in a clinical trial, showing better clearance of H. pylori in patients treated with NAC before antibiotic treatment.174 However, the exact mechanism underlying the reported therapeutic effect of NAC in the disruption of biofilms and overcoming H. pylori antibiotic resistance has yet to be defined. In a recent study, the combination of antibiotics and rhamnolipid, a glycolipid biosurfactant capable of disrupting biofilm and potentially inhibiting bacterial adhesion, was reported to effectively inhibit biofilm formation in vitro.175 Although the results are promising, the roles of rhamnolipid and anti-biofilm compounds need further investigation in vitro and in vivo.

Phage therapy

The rise in antibiotic resistance has increased interest in studying bacteriophages, particularly lytic bacteriophages. Phage therapy has various potential advantages over antibiotics because phages and their cleaved proteins are highly specific, affecting the target strain but not the microbiome.176 Furthermore, phages exclusively replicate at the site of infection, and no secondary effects have been reported.177 Few studies have investigated the presence of phages in H. pylori strains, including prophages and lytic phages.178179180181 However, the exact function and properties of phages to be used therapeutically are yet to be determined. Although phage therapy appears to be a promising approach for future treatment options for H. pylori infection, further investigations are required to improve our understanding of phage and H. pylori interactions, which are still in the exploratory stage.

Vaccination

Developing an effective vaccine against H. pylori could be a game-changer in preventing infections and reducing the reliance on antibiotic treatment. Several vaccine candidates are currently under investigation, aiming to elicit a protective immune response against H. pylori; however, few have shown a protective effect.182183184185186187188189190191192193194 In a randomized, double-blind, placebo-controlled, phase 3 clinical trial, three doses of oral recombinant H pylori vaccine were introduced in children and were followed up for the next three years.194 The vaccine was effective against H. pylori in 71.8% of subjects without any serious adverse events. However, the authors suggested a longer follow-up period to confirm protective effects against H. pylori-associated diseases. The search for an effective vaccine is in the exploratory stage and needs further investigations, considering better design of vaccine strategies, optimal combination of antigens, selection of suitable adjuvants, and proper delivery carriers. Additionally, given that H. pylori infection usually occurs in early childhood, the optimal timing of vaccination and subsequent follow-up strategies to ensure the protective effects of vaccination should be further elucidated.

CONCLUSIONS

The increasing rates of H. pylori antibiotic resistance and MDR strains pose significant challenges for eradication therapy. Substantial progress has been made in understanding the fundamental mechanisms underlying antibiotic resistance in H. pylori, including genetic mutations, efflux pump systems, and biofilm formation. To overcome antibiotic resistance of H. pylori, antibiotic stewardship and tailored therapy based on antimicrobial susceptibility testing are recommended. Strategies such as adjuvant treatment, phage therapy, and vaccine development are currently being explored. In addition to optimizing currently available treatment options, continuous monitoring of local resistance profiles, ongoing research, and the development of innovative therapies are required to effectively manage antibiotic-resistant H. pylori infections and associated gastrointestinal diseases.

Notes

Funding: This research was supported by National Research Foundation (NRF) grant funded by the Korean government (MIST) (NRF-2021R1C1C1010631) and Hallym University Research Fund.

Disclosure: The authors have no potential conflicts of interest to disclose.

Author Contributions:

  • Conceptualization: Gong EJ.

  • Funding acquisition: Gong EJ.

  • Investigation: Hasanuzzaman M, Bang CS.

  • Methodology: Gong EJ.

  • Project administration: Gong EJ.

  • Supervision: Gong EJ.

  • Writing - original draft: Hasanuzzaman M.

  • Writing - review & editing: Bang CS, Gong EJ.

References

1. Suerbaum S, Michetti P. Helicobacter pylori infection. N Engl J Med. 2002; 347(15):1175–1186. PMID: 12374879.
2. Kusters JG, van Vliet AH, Kuipers EJ. Pathogenesis of Helicobacter pylori infection. Clin Microbiol Rev. 2006; 19(3):449–490. PMID: 16847081.
3. Kayali S, Manfredi M, Gaiani F, Bianchi L, Bizzarri B, Leandro G, et al. Helicobacter pylori, transmission routes and recurrence of infection: state of the art. Acta Biomed. 2018; 89(8-S):72–76.
4. Yokota S, Konno M, Fujiwara S, Toita N, Takahashi M, Yamamoto S, et al. Intrafamilial, preferentially mother-to-child and intraspousal, Helicobacter pylori infection in Japan determined by mutilocus sequence typing and random amplified polymorphic DNA fingerprinting. Helicobacter. 2015; 20(5):334–342. PMID: 25664889.
5. McColl KE. Clinical practice. Helicobacter pylori infection. N Engl J Med. 2010; 362(17):1597–1604. PMID: 20427808.
6. Hooi JK, Lai WY, Ng WK, Suen MM, Underwood FE, Tanyingoh D, et al. Global prevalence of Helicobacter pylori infection: systematic review and meta-analysis. Gastroenterology. 2017; 153(2):420–429. PMID: 28456631.
7. Tshibangu-Kabamba E, Yamaoka Y. Helicobacter pylori infection and antibiotic resistance - from biology to clinical implications. Nat Rev Gastroenterol Hepatol. 2021; 18(9):613–629. PMID: 34002081.
8. Lee JH, Choi KD, Jung HY, Baik GH, Park JK, Kim SS, et al. Seroprevalence of Helicobacter pylori in Korea: a multicenter, nationwide study conducted in 2015 and 2016. Helicobacter. 2018; 23(2):e12463. PMID: 29345022.
9. Li Y, Choi H, Leung K, Jiang F, Graham DY, Leung WK. Global prevalence of Helicobacter pylori infection between 1980 and 2022: a systematic review and meta-analysis. Lancet Gastroenterol Hepatol. 2023; 8(6):553–564. PMID: 37086739.
10. Nagy P, Johansson S, Molloy-Bland M. Systematic review of time trends in the prevalence of Helicobacter pylori infection in China and the USA. Gut Pathog. 2016; 8(1):8. PMID: 26981156.
11. Malfertheiner P, Megraud F, Rokkas T, Gisbert JP, Liou JM, Schulz C, et al. Management of Helicobacter pylori infection: the Maastricht VI/Florence consensus report. Gut. 2022; 71(9):1724–1762.
12. Jung HK, Kang SJ, Lee YC, Yang HJ, Park SY, Shin CM, et al. Evidence-based guidelines for the treatment of Helicobacter pylori infection in Korea 2020. Gut Liver. 2021; 15(2):168–195. PMID: 33468712.
13. Dore MP, Lu H, Graham DY. Role of bismuth in improving Helicobacter pylori eradication with triple therapy. Gut. 2016; 65(5):870–878. PMID: 26848181.
14. Thung I, Aramin H, Vavinskaya V, Gupta S, Park JY, Crowe SE, et al. Review article: the global emergence of Helicobacter pylori antibiotic resistance. Aliment Pharmacol Ther. 2016; 43(4):514–533. PMID: 26694080.
15. Savoldi A, Carrara E, Graham DY, Conti M, Tacconelli E. Prevalence of antibiotic resistance in Helicobacter pylori: a systematic review and meta-analysis in World Health Organization Regions. Gastroenterology. 2018; 155(5):1372–1382.e17. PMID: 29990487.
16. Lee JW, Kim N, Choi SI, Jang JY, Song CH, Nam RH, et al. Prevalence and trends of multiple antimicrobial resistance of Helicobacter pylori in one tertiary hospital for 20 years in Korea. Helicobacter. 2023; 28(1):e12939. PMID: 36478622.
17. Ho JJ, Navarro M, Sawyer K, Elfanagely Y, Moss SF. Helicobacter pylori antibiotic resistance in the united states between 2011 and 2021: a systematic review and meta-analysis. Am J Gastroenterol. 2022; 117(8):1221–1230. PMID: 35509128.
18. Kim SE, Park MI, Park SJ, Moon W, Choi YJ, Cheon JH, et al. Trends in Helicobacter pylori eradication rates by first-line triple therapy and related factors in eradication therapy. Korean J Intern Med. 2015; 30(6):801–807. PMID: 26552455.
19. Gong EJ, Yun SC, Jung HY, Lim H, Choi KS, Ahn JY, et al. Meta-analysis of first-line triple therapy for Helicobacter pylori eradication in Korea: is it time to change? J Korean Med Sci. 2014; 29(5):704–713. PMID: 24851029.
20. Choe Y. Antibiotic resistance and Helicobacter pylori eradication therapy. Korean J Helicobacter Up Gastrointest Res. 2023; 23(3):218–221.
21. Megraud F, Bruyndonckx R, Coenen S, Wittkop L, Huang TD, Hoebeke M, et al. Helicobacter pylori resistance to antibiotics in Europe in 2018 and its relationship to antibiotic consumption in the community. Gut. 2021; 70(10):1815–1822. PMID: 33837118.
22. Shin WG, Lee SW, Baik GH, Huh KC, Lee SI, Chung JW, et al. Eradication rates of Helicobacter pylori in Korea over the past 10 years and correlation of the amount of antibiotics use: nationwide survey. Helicobacter. 2016; 21(4):266–278. PMID: 26470999.
23. Boltin D, Levi Z, Gingold-Belfer R, Gabay H, Shochat T, Niv Y, et al. Impact of previous exposure to macrolide antibiotics on Helicobacter pylori infection treatment outcomes. Am J Gastroenterol. 2019; 114(6):900–906. PMID: 31095531.
24. Tacconelli E, Carrara E, Savoldi A, Harbarth S, Mendelson M, Monnet DL, et al. Discovery, research, and development of new antibiotics: the WHO priority list of antibiotic-resistant bacteria and tuberculosis. Lancet Infect Dis. 2018; 18(3):318–327. PMID: 29276051.
25. Gong EJ, Ahn JY, Kim JM, Lee SM, Na HK, Lee JH, et al. Genotypic and phenotypic resistance to clarithromycin in Helicobacter pylori strains. J Clin Med. 2020; 9(6):1930. PMID: 32575584.
26. Tuan VP, Narith D, Tshibangu-Kabamba E, Dung HD, Viet PT, Sokomoth S, et al. A next-generation sequencing-based approach to identify genetic determinants of antibiotic resistance in Cambodian Helicobacter pylori clinical isolates. J Clin Med. 2019; 8(6):858. PMID: 31208076.
27. Lauener FN, Imkamp F, Lehours P, Buissonnière A, Benejat L, Zbinden R, et al. Genetic determinants and prediction of antibiotic resistance phenotypes in Helicobacter pylori . J Clin Med. 2019; 8(1):53. PMID: 30621024.
28. Aldred KJ, Kerns RJ, Osheroff N. Mechanism of quinolone action and resistance. Biochemistry. 2014; 53(10):1565–1574. PMID: 24576155.
29. Zhang Z, Liu ZQ, Zheng PY, Tang FA, Yang PC. Influence of efflux pump inhibitors on the multidrug resistance of Helicobacter pylori . World J Gastroenterol. 2010; 16(10):1279–1284. PMID: 20222174.
30. Mégraud F. H pylori antibiotic resistance: prevalence, importance, and advances in testing. Gut. 2004; 53(9):1374–1384. PMID: 15306603.
31. Yonezawa H, Osaki T, Kamiya S. Biofilm formation by Helicobacter pylori and its involvement for antibiotic resistance. BioMed Res Int. 2015; 2015:914791. PMID: 26078970.
32. Kadkhodaei S, Siavoshi F, Akbari Noghabi K. Mucoid and coccoid Helicobacter pylori with fast growth and antibiotic resistance. Helicobacter. 2020; 25(2):e12678. PMID: 31880001.
33. Dore MP, Leandro G, Realdi G, Sepulveda AR, Graham DY. Effect of pretreatment antibiotic resistance to metronidazole and clarithromycin on outcome of Helicobacter pylori therapy: a meta-analytical approach. Dig Dis Sci. 2000; 45(1):68–76. PMID: 10695616.
34. Binh TT, Shiota S, Suzuki R, Matsuda M, Trang TT, Kwon DH, et al. Discovery of novel mutations for clarithromycin resistance in Helicobacter pylori by using next-generation sequencing. J Antimicrob Chemother. 2014; 69(7):1796–1803. PMID: 24648504.
35. Tshibangu-Kabamba E, Ngoma-Kisoko PJ, Tuan VP, Matsumoto T, Akada J, Kido Y, et al. Next-generation sequencing of the whole bacterial genome for tracking molecular insight into the broad-spectrum antimicrobial resistance of Helicobacter pylori clinical isolates from the Democratic Republic of Congo. Microorganisms. 2020; 8(6):8.
36. Domanovich-Asor T, Motro Y, Khalfin B, Craddock HA, Peretz A, Moran-Gilad J. Genomic analysis of antimicrobial resistance genotype-to-phenotype agreement in Helicobacter pylori . Microorganisms. 2020; 9(1):2. PMID: 33374988.
37. Nakamura M, Spiller RC, Barrett DA, Wibawa JI, Kumagai N, Tsuchimoto K, et al. Gastric juice, gastric tissue and blood antibiotic concentrations following omeprazole, amoxicillin and clarithromycin triple therapy. Helicobacter. 2003; 8(4):294–299. PMID: 12950601.
38. Zullo A. The current role of dual therapy for treatment of Helicobacter pylori: back to the future? Eur J Gastroenterol Hepatol. 2020; 32(5):555–556. PMID: 31922977.
39. Gerrits MM, van Vliet AH, Kuipers EJ, Kusters JG. Helicobacter pylori and antimicrobial resistance: molecular mechanisms and clinical implications. Lancet Infect Dis. 2006; 6(11):699–709. PMID: 17067919.
40. Wilke MS, Lovering AL, Strynadka NC. Beta-lactam antibiotic resistance: a current structural perspective. Curr Opin Microbiol. 2005; 8(5):525–533. PMID: 16129657.
41. Kuo YT, Liou JM, El-Omar EM, Wu JY, Leow AH, Goh KL, et al. Primary antibiotic resistance in Helicobacter pylori in the Asia-Pacific region: a systematic review and meta-analysis. Lancet Gastroenterol Hepatol. 2017; 2(10):707–715. PMID: 28781119.
42. Gong EJ, Ahn JY. Antimicrobial resistance of Helicobacter pylori isolates in Korea. Korean J Helicobacter Up Gastrointest Res. 2018; 18(2):82–88.
43. Cosme A, Torrente Iranzo S, Montes Ros M, Fernández-Reyes Silvestre M, Alonso Galán H, Lizasoain J, et al. Helicobacter pylori antimicrobial resistance during a 5-year period (2013-2017) in northern Spain and its relationship with the eradication therapies. Helicobacter. 2019; 24(1):e12557. PMID: 30460730.
44. Bujanda L, Nyssen OP, Vaira D, Saracino IM, Fiorini G, Lerang F, et al. Antibiotic resistance prevalence and trends in patients infected with Helicobacter pylori in the period 2013-2020: results of the European Registry on H. pylori Management (Hp-EuReg). Antibiotics (Basel). 2021; 10(9):1058. PMID: 34572640.
45. Gerrits MM, Godoy AP, Kuipers EJ, Ribeiro ML, Stoof J, Mendonça S, et al. Multiple mutations in or adjacent to the conserved penicillin-binding protein motifs of the penicillin-binding protein 1A confer amoxicillin resistance to Helicobacter pylori . Helicobacter. 2006; 11(3):181–187. PMID: 16684266.
46. Kwon YH, Kim JY, Kim N, Park JH, Nam RH, Lee SM, et al. Specific mutations of penicillin-binding protein 1A in 77 clinically acquired amoxicillin-resistant Helicobacter pylori strains in comparison with 77 amoxicillin-susceptible strains. Helicobacter. 2017; 22(6):e12437.
47. Kim JM, Kim JS, Kim N, Kim SG, Jung HC, Song IS. Comparison of primary and secondary antimicrobial minimum inhibitory concentrations for Helicobacter pylori isolated from Korean patients. Int J Antimicrob Agents. 2006; 28(1):6–13. PMID: 16750904.
48. Kim BJ, Kim JG. Substitutions in penicillin-binding protein 1 in amoxicillin-resistant Helicobacter pylori strains isolated from Korean patients. Gut Liver. 2013; 7(6):655–660. PMID: 24312705.
49. Rimbara E, Noguchi N, Kawai T, Sasatsu M. Mutations in penicillin-binding proteins 1, 2 and 3 are responsible for amoxicillin resistance in Helicobacter pylori . J Antimicrob Chemother. 2008; 61(5):995–998. PMID: 18276599.
50. Tseng YS, Wu DC, Chang CY, Kuo CH, Yang YC, Jan CM, et al. Amoxicillin resistance with beta-lactamase production in Helicobacter pylori . Eur J Clin Invest. 2009; 39(9):807–812. PMID: 19614952.
51. Co EM, Schiller NL. Resistance mechanisms in an in vitro-selected amoxicillin-resistant strain of Helicobacter pylori . Antimicrob Agents Chemother. 2006; 50(12):4174–4176. PMID: 17000747.
52. Qureshi NN, Gallaher B, Schiller NL. Evolution of amoxicillin resistance of Helicobacter pylori in vitro: characterization of resistance mechanisms. Microb Drug Resist. 2014; 20(6):509–516. PMID: 24901497.
53. Park SY, Lee EH, Kim D, Song YG, Jeong SJ. Novel mutations conferring amoxicillin resistance in Helicobacter pylori in South Korea. Antibiotics (Basel). 2023; 12(4):748. PMID: 37107110.
54. Rodvold KA. Clinical pharmacokinetics of clarithromycin. Clin Pharmacokinet. 1999; 37(5):385–398. PMID: 10589373.
55. Erah PO, Goddard AF, Barrett DA, Shaw PN, Spiller RC. The stability of amoxycillin, clarithromycin and metronidazole in gastric juice: relevance to the treatment of Helicobacter pylori infection. J Antimicrob Chemother. 1997; 39(1):5–12. PMID: 9044021.
56. Versalovic J, Shortridge D, Kibler K, Griffy MV, Beyer J, Flamm RK, et al. Mutations in 23S rRNA are associated with clarithromycin resistance in Helicobacter pylori . Antimicrob Agents Chemother. 1996; 40(2):477–480. PMID: 8834903.
57. Taylor DE, Ge Z, Purych D, Lo T, Hiratsuka K. Cloning and sequence analysis of two copies of a 23S rRNA gene from Helicobacter pylori and association of clarithromycin resistance with 23S rRNA mutations. Antimicrob Agents Chemother. 1997; 41(12):2621–2628. PMID: 9420030.
58. Lyu T, Cheung KS, Deng Z, Ni L, Chen C, Wu J, et al. Whole genome sequencing reveals novel genetic mutations of Helicobacter pylori associating with resistance to clarithromycin and levofloxacin. Helicobacter. 2023; 28(4):e12972. PMID: 36965192.
59. Hirata K, Suzuki H, Nishizawa T, Tsugawa H, Muraoka H, Saito Y, et al. Contribution of efflux pumps to clarithromycin resistance in Helicobacter pylori . J Gastroenterol Hepatol. 2010; 25(Suppl 1):S75–S79. PMID: 20586871.
60. Choi JH, Yang YJ, Bang CS, Lee JJ, Baik GH. Current status of the third-line Helicobacter pylori eradication. Gastroenterol Res Pract. 2018; 2018:6523653. PMID: 29853863.
61. Liou JM, Lee YC, Wu MS. Taiwan Gastrointestinal Disease and Helicobacter Consortium. Treatment of refractory Helicobacter pylori infection-tailored or empirical therapy. Gut Liver. 2022; 16(1):8–18. PMID: 33782215.
62. Redgrave LS, Sutton SB, Webber MA, Piddock LJ. Fluoroquinolone resistance: mechanisms, impact on bacteria, and role in evolutionary success. Trends Microbiol. 2014; 22(8):438–445. PMID: 24842194.
63. Yoshida H, Bogaki M, Nakamura M, Nakamura S. Quinolone resistance-determining region in the DNA gyrase gyrA gene of Escherichia coli . Antimicrob Agents Chemother. 1990; 34(6):1271–1272. PMID: 2168148.
64. Yoshida H, Bogaki M, Nakamura M, Yamanaka LM, Nakamura S. Quinolone resistance-determining region in the DNA gyrase gyrB gene of Escherichia coli . Antimicrob Agents Chemother. 1991; 35(8):1647–1650. PMID: 1656869.
65. Lee JW, Kim N, Nam RH, Park JH, Kim JM, Jung HC, et al. Mutations of Helicobacter pylori associated with fluoroquinolone resistance in Korea. Helicobacter. 2011; 16(4):301–310. PMID: 21762270.
66. Rimbara E, Noguchi N, Kawai T, Sasatsu M. Fluoroquinolone resistance in Helicobacter pylori: role of mutations at position 87 and 91 of GyrA on the level of resistance and identification of a resistance conferring mutation in GyrB. Helicobacter. 2012; 17(1):36–42.
67. Miyachi H, Miki I, Aoyama N, Shirasaka D, Matsumoto Y, Toyoda M, et al. Primary levofloxacin resistance and gyrA/B mutations among Helicobacter pylori in Japan. Helicobacter. 2006; 11(4):243–249. PMID: 16882327.
68. Debets-Ossenkopp YJ, Namavar F, MacLaren DM. Effect of an acidic environment on the susceptibility of Helicobacter pylori to trospectomycin and other antimicrobial agents. Eur J Clin Microbiol Infect Dis. 1995; 14(4):353–355. PMID: 7649202.
69. Dingsdag SA, Hunter N. Metronidazole: an update on metabolism, structure-cytotoxicity and resistance mechanisms. J Antimicrob Chemother. 2018; 73(2):265–279. PMID: 29077920.
70. Martínez-Júlvez M, Rojas AL, Olekhnovich I, Espinosa Angarica V, Hoffman PS, Sancho J. Structure of RdxA--an oxygen-insensitive nitroreductase essential for metronidazole activation in Helicobacter pylori. FEBS J. 2012; 279(23):4306–4317. PMID: 23039228.
71. Gong Y, Zhai K, Sun L, He L, Wang H, Guo Y, et al. RdxA diversity and mutations associated with metronidazole resistance of Helicobacter pylori . Microbiol Spectr. 2023; 11(2):e0390322. PMID: 36943041.
72. Lee SM, Kim N, Kwon YH, Nam RH, Kim JM, Park JY, et al. rdxA, frxA, and efflux pump in metronidazole-resistant Helicobacter pylori: their relation to clinical outcomes. J Gastroenterol Hepatol. 2018; 33(3):681–688. PMID: 28748532.
73. Goodwin A, Kersulyte D, Sisson G, Veldhuyzen van Zanten SJ, Berg DE, Hoffman PS. Metronidazole resistance in Helicobacter pylori is due to null mutations in a gene (rdxA) that encodes an oxygen-insensitive NADPH nitroreductase. Mol Microbiol. 1998; 28(2):383–393. PMID: 9622362.
74. Jeong JY, Mukhopadhyay AK, Dailidiene D, Wang Y, Velapatiño B, Gilman RH, et al. Sequential inactivation of rdxA (HP0954) and frxA (HP0642) nitroreductase genes causes moderate and high-level metronidazole resistance in Helicobacter pylori . J Bacteriol. 2000; 182(18):5082–5090. PMID: 10960091.
75. Binh TT, Suzuki R, Trang TT, Kwon DH, Yamaoka Y. Search for novel candidate mutations for metronidazole resistance in Helicobacter pylori using next-generation sequencing. Antimicrob Agents Chemother. 2015; 59(4):2343–2348. PMID: 25645832.
76. Albert TJ, Dailidiene D, Dailide G, Norton JE, Kalia A, Richmond TA, et al. Mutation discovery in bacterial genomes: metronidazole resistance in Helicobacter pylori . Nat Methods. 2005; 2(12):951–953. PMID: 16299480.
77. Kwon DH, Kato M, El-Zaatari FA, Osato MS, Graham DY. Frame-shift mutations in NAD(P)H flavin oxidoreductase encoding gene (frxA) from metronidazole resistant Helicobacter pylori ATCC43504 and its involvement in metronidazole resistance. FEMS Microbiol Lett. 2000; 188(2):197–202. PMID: 10913705.
78. Kim SY, Joo YM, Lee HS, Chung IS, Yoo YJ, Merrell DS, et al. Genetic analysis of Helicobacter pylori clinical isolates suggests resistance to metronidazole can occur without the loss of functional rdxA. J Antibiot (Tokyo). 2009; 62(1):43–50. PMID: 19132060.
79. Zhang S, Wang X, Wise MJ, He Y, Chen H, Liu A, et al. Mutations of Helicobacter pylori RdxA are mainly related to the phylogenetic origin of the strain and not to metronidazole resistance. J Antimicrob Chemother. 2020; 75(11):3152–3155. PMID: 32676634.
80. An B, Moon BS, Lim HC, Lee YC, Kim H, Lee G, et al. Analysis of gene mutations associated with antibiotic resistance in Helicobacter pylori strains isolated from Korean patients. Korean J Helicobacter Up Gastrointest Res. 2014; 14(2):95–102.
81. Lee SY, Lee YC, Pyo JH, Kwon DH, Rhee JC, Kim JJ. Quasispecies of antibacterial heteroresistant Helicobacter pylori . Korean J Helicobacter Up Gastrointest Res. 2004; 4(1):7–14.
82. Kwon DH, El-Zaatari FA, Kato M, Osato MS, Reddy R, Yamaoka Y, et al. Analysis of rdxA and involvement of additional genes encoding NAD(P)H flavin oxidoreductase (FrxA) and ferredoxin-like protein (FdxB) in metronidazole resistance of Helicobacter pylori . Antimicrob Agents Chemother. 2000; 44(8):2133–2142. PMID: 10898687.
83. Tsugawa H, Suzuki H, Satoh K, Hirata K, Matsuzaki J, Saito Y, et al. Two amino acids mutation of ferric uptake regulator determines Helicobacter pylori resistance to metronidazole. Antioxid Redox Signal. 2011; 14(1):15–23. PMID: 20518707.
84. Attaran B, Falsafi T, Ghorbanmehr N. Effect of biofilm formation by clinical isolates of Helicobacter pylori on the efflux-mediated resistance to commonly used antibiotics. World J Gastroenterol. 2017; 23(7):1163–1170. PMID: 28275296.
85. Graham DY. Antibiotic resistance in Helicobacter pylori: implications for therapy. Gastroenterology. 1998; 115(5):1272–1277. PMID: 9797384.
86. Chopra I, Roberts M. Tetracycline antibiotics: mode of action, applications, molecular biology, and epidemiology of bacterial resistance. Microbiol Mol Biol Rev. 2001; 65(2):232–260. PMID: 11381101.
87. Lee JH, Ahn JY, Choi KD, Jung HY, Kim JM, Baik GH, et al. Nationwide antibiotic resistance mapping of Helicobacter pylori in Korea: a prospective multicenter study. Helicobacter. 2019; 24(4):e12592. PMID: 31111572.
88. Dailidiene D, Bertoli MT, Miciuleviciene J, Mukhopadhyay AK, Dailide G, Pascasio MA, et al. Emergence of tetracycline resistance in Helicobacter pylori: multiple mutational changes in 16S ribosomal DNA and other genetic loci. Antimicrob Agents Chemother. 2002; 46(12):3940–3946. PMID: 12435699.
89. Gerrits MM, de Zoete MR, Arents NL, Kuipers EJ, Kusters JG. 16S rRNA mutation-mediated tetracycline resistance in Helicobacter pylori . Antimicrob Agents Chemother. 2002; 46(9):2996–3000. PMID: 12183259.
90. Trieber CA, Taylor DE. Mutations in the 16S rRNA genes of Helicobacter pylori mediate resistance to tetracycline. J Bacteriol. 2002; 184(8):2131–2140. PMID: 11914344.
91. Wu JY, Kim JJ, Reddy R, Wang WM, Graham DY, Kwon DH. Tetracycline-resistant clinical Helicobacter pylori isolates with and without mutations in 16S rRNA-encoding genes. Antimicrob Agents Chemother. 2005; 49(2):578–583. PMID: 15673736.
92. Rothstein DM. Rifamycins, alone and in combination. Cold Spring Harb Perspect Med. 2016; 6(7):a027011. PMID: 27270559.
93. Crabol Y, Catherinot E, Veziris N, Jullien V, Lortholary O. Rifabutin: where do we stand in 2016? J Antimicrob Chemother. 2016; 71(7):1759–1771. PMID: 27009031.
94. Gisbert JP, Calvet X. Review article: rifabutin in the treatment of refractory Helicobacter pylori infection. Aliment Pharmacol Ther. 2012; 35(2):209–221. PMID: 22129228.
95. Hays C, Burucoa C, Lehours P, Tran CT, Leleu A, Raymond J. Molecular characterization of Helicobacter pylori resistance to rifamycins. Helicobacter. 2018; 23(1):e12451.
96. Choi YI, Jeong SH, Chung JW, Park DK, Kim KO, Kwon KA, et al. Rifabutin and furazolidone could be the candidates of the rescue regimen for antibiotic-resistant H. pylori in Korea. Can J Infect Dis Med Microbiol. 2019; 2019:9351801. PMID: 31360270.
97. Heep M, Beck D, Bayerdörffer E, Lehn N. Rifampin and rifabutin resistance mechanism in Helicobacter pylori . Antimicrob Agents Chemother. 1999; 43(6):1497–1499. PMID: 10348780.
98. Nishizawa T, Suzuki H, Matsuzaki J, Muraoka H, Tsugawa H, Hirata K, et al. Helicobacter pylori resistance to rifabutin in the last 7 years. Antimicrob Agents Chemother. 2011; 55(11):5374–5375. PMID: 21896915.
99. Heep M, Lehn N, Brandstätter B, Rieger U, Senzenberger S, Wehrl W. Detection of rifabutin resistance and association of rpoB mutations with resistance to four rifamycin derivatives in Helicobacter pylori . Eur J Clin Microbiol Infect Dis. 2002; 21(2):143–145. PMID: 11939398.
100. Suzuki S, Suzuki H, Nishizawa T, Kaneko F, Ootani S, Muraoka H, et al. Past rifampicin dosing determines rifabutin resistance of Helicobacter pylori . Digestion. 2009; 79(1):1–4. PMID: 19142036.
101. Mori H, Suzuki H, Matsuzaki J, Tsugawa H, Fukuhara S, Miyoshi S, et al. Rifabutin-based 10-day and 14-day triple therapy as a third-line and fourth-line regimen for Helicobacter pylori eradication: a pilot study. United European Gastroenterol J. 2016; 4(3):380–387.
102. Tsugawa H, Suzuki H, Muraoka H, Ikeda F, Hirata K, Matsuzaki J, et al. Enhanced bacterial efflux system is the first step to the development of metronidazole resistance in Helicobacter pylori . Biochem Biophys Res Commun. 2011; 404(2):656–660. PMID: 21147064.
103. Ge X, Cai Y, Chen Z, Gao S, Geng X, Li Y, et al. Bifunctional enzyme SpoT Is involved in biofilm formation of Helicobacter pylori with multidrug resistance by upregulating efflux pump Hp1174 (gluP). Antimicrob Agents Chemother. 2018; 62(11):e00957-18. PMID: 30181372.
104. Liu ZQ, Zheng PY, Yang PC. Efflux pump gene hefA of Helicobacter pylori plays an important role in multidrug resistance. World J Gastroenterol. 2008; 14(33):5217–5222. PMID: 18777600.
105. Cai Y, Wang C, Chen Z, Xu Z, Li H, Li W, et al. Transporters HP0939, HP0497, and HP0471 participate in intrinsic multidrug resistance and biofilm formation in Helicobacter pylori by enhancing drug efflux. Helicobacter. 2020; 25(4):e12715. PMID: 32548895.
106. Singh S, Singh SK, Chowdhury I, Singh R. Understanding the mechanism of bacterial biofilms resistance to antimicrobial agents. Open Microbiol J. 2017; 11(1):53–62. PMID: 28553416.
107. Flemming HC, Wingender J, Szewzyk U, Steinberg P, Rice SA, Kjelleberg S. Biofilms: an emergent form of bacterial life. Nat Rev Microbiol. 2016; 14(9):563–575. PMID: 27510863.
108. Koo H, Allan RN, Howlin RP, Stoodley P, Hall-Stoodley L. Targeting microbial biofilms: current and prospective therapeutic strategies. Nat Rev Microbiol. 2017; 15(12):740–755. PMID: 28944770.
109. Yonezawa H, Osaki T, Kurata S, Zaman C, Hanawa T, Kamiya S. Assessment of in vitro biofilm formation by Helicobacter pylori . J Gastroenterol Hepatol. 2010; 25(Suppl 1):S90–S94. PMID: 20586874.
110. Carron MA, Tran VR, Sugawa C, Coticchia JM. Identification of Helicobacter pylori biofilms in human gastric mucosa. J Gastrointest Surg. 2006; 10(5):712–717. PMID: 16713544.
111. Cole SP, Harwood J, Lee R, She R, Guiney DG. Characterization of monospecies biofilm formation by Helicobacter pylori . J Bacteriol. 2004; 186(10):3124–3132. PMID: 15126474.
112. Wong EH, Ng CG, Chua EG, Tay AC, Peters F, Marshall BJ, et al. Comparative genomics revealed multiple Helicobacter pylori genes associated with biofilm formation in vitro. PLoS One. 2016; 11(11):e0166835. PMID: 27870886.
113. Bode G, Mauch F, Malfertheiner P. The coccoid forms of Helicobacter pylori. Criteria for their viability. Epidemiol Infect. 1993; 111(3):483–490. PMID: 8270008.
114. Kusters JG, Gerrits MM, Van Strijp JA, Vandenbroucke-Grauls CM. Coccoid forms of Helicobacter pylori are the morphologic manifestation of cell death. Infect Immun. 1997; 65(9):3672–3679. PMID: 9284136.
115. El-Halfawy OM, Valvano MA. Antimicrobial heteroresistance: an emerging field in need of clarity. Clin Microbiol Rev. 2015; 28(1):191–207. PMID: 25567227.
116. Kocsmár É, Kocsmár I, Buzás GM, Szirtes I, Wacha J, Takáts A, et al. Helicobacter pylori heteroresistance to clarithromycin in adults-new data by in situ detection and improved concept. Helicobacter. 2020; 25(1):e12670. PMID: 31701608.
117. Kim JJ, Kim JG, Kwon DH. Mixed-infection of antibiotic susceptible and resistant Helicobacter pylori isolates in a single patient and underestimation of antimicrobial susceptibility testing. Helicobacter. 2003; 8(3):202–206. PMID: 12752732.
118. Lee SY, Kim JJ, Kwon DH. Synchronous infection of heteroresistant Helicobacter pylori . Korean J Helicobacter Res Pract. 2003; 3(2):110–116.
119. Kao CY, Lee AY, Huang AH, Song PY, Yang YJ, Sheu SM, et al. Heteroresistance of Helicobacter pylori from the same patient prior to antibiotic treatment. Infect Genet Evol. 2014; 23:196–202. PMID: 24576534.
120. Arévalo-Jaimes BV, Rojas-Rengifo DF, Jaramillo CA, de Molano BM, Vera-Chamorro JF, Del Pilar Delgado M. Genotypic determination of resistance and heteroresistance to clarithromycin in Helicobacter pylori isolates from antrum and corpus of Colombian symptomatic patients. BMC Infect Dis. 2019; 19(1):546. PMID: 31226948.
121. van der Ende A, van Doorn LJ, Rooijakkers S, Feller M, Tytgat GN, Dankert J. Clarithromycin-susceptible and -resistant Helicobacter pylori isolates with identical randomly amplified polymorphic DNA-PCR genotypes cultured from single gastric biopsy specimens prior to antibiotic therapy. J Clin Microbiol. 2001; 39(7):2648–2651. PMID: 11427586.
122. Keikha M, Karbalaei M. Prevalence of antibiotic heteroresistance associated with Helicobacter pylori infection: a systematic review and meta-analysis. Microb Pathog. 2022; 170:105720. PMID: 35964816.
123. Li H, Shen Y, Song X, Tang X, Hu R, Marshall BJ, et al. Need for standardization and harmonization of Helicobacter pylori antimicrobial susceptibility testing. Helicobacter. 2022; 27(2):e12873. PMID: 35151236.
124. Kim YJ, Chung WC. Eradication therapy for Helicobacter pylori with diagnostic test for clarithromycin resistance. Korean J Helicobacter Up Gastrointest Res. 2019; 19(4):225–230.
125. Woo HY, Park DI, Park H, Kim MK, Kim DH, Kim IS, et al. Dual-priming oligonucleotide-based multiplex PCR for the detection of Helicobacter pylori and determination of clarithromycin resistance with gastric biopsy specimens. Helicobacter. 2009; 14(1):22–28. PMID: 19191892.
126. Redondo JJ, Keller PM, Zbinden R, Wagner K. A novel RT-PCR for the detection of Helicobacter pylori and identification of clarithromycin resistance mediated by mutations in the 23S rRNA gene. Diagn Microbiol Infect Dis. 2018; 90(1):1–6. PMID: 29111147.
127. Binmaeil H, Hanafiah A, Mohamed Rose I, Raja Ali RA. Development and validation of multiplex quantitative PCR assay for detection of Helicobacter pylori and mutations conferring resistance to clarithromycin and levofloxacin in gastric biopsy. Infect Drug Resist. 2021; 14:4129–4145. PMID: 34675558.
128. Lehours P, Siffré E, Mégraud F. DPO multiplex PCR as an alternative to culture and susceptibility testing to detect Helicobacter pylori and its resistance to clarithromycin. BMC Gastroenterol. 2011; 11(1):112. PMID: 22004003.
129. Nahm JH, Kim WK, Kwon Y, Kim H. Detection of Helicobacter pylori with clarithromycin resistance-associated mutations using peptide nucleic acid probe-based melting point analysis. Helicobacter. 2019; 24(5):e12634. PMID: 31304664.
130. Cambau E, Allerheiligen V, Coulon C, Corbel C, Lascols C, Deforges L, et al. Evaluation of a new test, genotype HelicoDR, for molecular detection of antibiotic resistance in Helicobacter pylori . J Clin Microbiol. 2009; 47(11):3600–3607. PMID: 19759218.
131. Lee JW, Kim N, Nam RH, Park JH, Choi YJ, Kim JM, et al. GenoType HelicoDR test in the determination of antimicrobial resistance of Helicobacter pylori in Korea. Scand J Gastroenterol. 2014; 49(9):1058–1067. PMID: 24957849.
132. Schabereiter-Gurtner C, Hirschl AM, Dragosics B, Hufnagl P, Puz S, Kovách Z, et al. Novel real-time PCR assay for detection of Helicobacter pylori infection and simultaneous clarithromycin susceptibility testing of stool and biopsy specimens. J Clin Microbiol. 2004; 42(10):4512–4518. PMID: 15472302.
133. Jehanne Q, Bénéjat L, Mégraud F, Bessède E, Lehours P. Evaluation of the Allplex™ H pylori and ClariR PCR Assay for Helicobacter pylori detection on gastric biopsies. Helicobacter. 2020; 25(4):e12702. PMID: 32426894.
134. Kim I, Maeng LS, Kim JS, Kim BW, Cheung DY, Kim JI, et al. Quantitative multiplex real-time polymerase chain reaction assay for the detection of Helicobacter pylori and clarithromycin resistance. BMC Microbiol. 2023; 23(1):155. PMID: 37237357.
135. Van den Poel B, Gils S, Micalessi I, Carton S, Christiaens P, Cuyle PJ, et al. Molecular detection of Helicobacter pylori and clarithromycin resistance in gastric biopsies: a prospective evaluation of RIDA®GENE Helicobacter pylori assay. Acta Clin Belg. 2021; 76(3):177–183. PMID: 31662122.
136. Park CG, Kim S, Jeon HS, Han S. Validation of loop-mediated isothermal amplification to detect Helicobacter pylori and 23S rRNA mutations: a prospective, observational clinical cohort study. J Clin Lab Anal. 2021; 35(1):e23563. PMID: 32893424.
137. Yari F, Abiri R, Aryan E, Ahmadi Jouybari T, Navabi J, Alvandi A. Loop-mediated isothermal amplification as a fast noninvasive method of Helicobacter pylori diagnosis. J Clin Lab Anal. 2016; 30(5):464–470. PMID: 26351213.
138. Kakiuchi T, Okuda M, Matsuo M, Fujimoto K. Smart Gene™ as an effective non-invasive point-of-care test to detect Helicobacter pylori clarithromycin-resistant mutation. J Gastroenterol Hepatol. 2022; 37(9):1719–1725. PMID: 35562319.
139. Liu M, Zhu P, Zhang L, Gong Y, Wang C, Sun L, et al. Single-cell identification, drug susceptibility test, and whole-genome sequencing of Helicobacter pylori directly from gastric biopsy by clinical antimicrobial susceptibility test Ramanometry. Clin Chem. 2022; 68(8):1064–1074. PMID: 35714147.
140. Saruuljavkhlan B, Yamaoka Y. Benefits of a molecular-based method for the detection of clarithromycin-resistant Helicobacter pylori . Gut Liver. 2021; 15(4):487–489. PMID: 34261816.
141. Hulten KG, Genta RM, Kalfus IN, Zhou Y, Zhang H, Graham DY. Comparison of culture with antibiogram to next-generation sequencing using bacterial isolates and formalin-fixed, paraffin-embedded gastric biopsies. Gastroenterology. 2021; 161(5):1433–1442.e2. PMID: 34293298.
142. Graham DY. Helicobacter pylori update: gastric cancer, reliable therapy, and possible benefits. Gastroenterology. 2015; 148(4):719–731.e3. PMID: 25655557.
143. Zou Y, Qian X, Liu X, Song Y, Song C, Wu S, et al. The effect of antibiotic resistance on Helicobacter pylori eradication efficacy: a systematic review and meta-analysis. Helicobacter. 2020; 25(4):e12714. PMID: 32533599.
144. Fallone CA, Moss SF, Malfertheiner P. Reconciliation of recent Helicobacter pylori treatment guidelines in a time of increasing resistance to antibiotics. Gastroenterology. 2019; 157(1):44–53. PMID: 30998990.
145. Shiotani A, Roy P, Lu H, Graham DY. Helicobacter pylori diagnosis and therapy in the era of antimicrobial stewardship. Therap Adv Gastroenterol. 2021; 14:17562848211064080.
146. Kim N, Kim JM, Kim CH, Park YS, Lee DH, Kim JS, et al. Institutional difference of antibiotic resistance of Helicobacter pylori strains in Korea. J Clin Gastroenterol. 2006; 40(8):683–687. PMID: 16940878.
147. Ko SW, Kim YJ, Chung WC, Lee SJ. Bismuth supplements as the first-line regimen for Helicobacter pylori eradication therapy: systemic review and meta-analysis. Helicobacter. 2019; 24(2):e12565. PMID: 30698318.
148. Wenzhen Y, Yumin L, Quanlin G, Kehu Y, Lei J, Donghai W, et al. Is antimicrobial susceptibility testing necessary before first-line treatment for Helicobacter pylori infection? Meta-analysis of randomized controlled trials. Intern Med. 2010; 49(12):1103–1109. PMID: 20558925.
149. Bae JH, Jo HH, Kwon JG, Kim EY. Efficacy of 7-day tailored therapy for Helicobacter pylori eradication based on clarithromycin resistance. Korean J Gastroenterol. 2023; 82(1):10–17. PMID: 37489077.
150. Cho SH, Park MS, Park SY, Kim DH, You HS, Kim HS. Effectiveness of 7-day triple therapy with half-dose clarithromycin for the eradication of Helicobacter pylori without the A2143G and A2142G point mutations of the 23S rRNA gene in a high clarithromycin resistance area. Front Med (Lausanne). 2023; 10:1150396. PMID: 37035320.
151. Nyssen OP, Espada M, Gisbert JP. Empirical vs. susceptibility-guided treatment of Helicobacter pylori infection: a systematic review and meta-analysis. Front Microbiol. 2022; 13:913436. PMID: 35774456.
152. López-Góngora S, Puig I, Calvet X, Villoria A, Baylina M, Muñoz N, et al. Systematic review and meta-analysis: susceptibility-guided versus empirical antibiotic treatment for Helicobacter pylori infection. J Antimicrob Chemother. 2015; 70(9):2447–2455. PMID: 26078393.
153. Gingold-Belfer R, Niv Y, Schmilovitz-Weiss H, Levi Z, Boltin D. Susceptibility-guided versus empirical treatment for Helicobacter pylori infection: a systematic review and meta-analysis. J Gastroenterol Hepatol. 2021; 36(10):2649–2658. PMID: 34114673.
154. Du RC, Ouyang YB, Lu NH, Hu Y. Research trends on vonoprazan-based therapy for Helicobacter pylori eradication: a bibliometric analysis from 2015 to 2023. Helicobacter. 2023; 28(5):e13012. PMID: 37515414.
155. Sugimoto M, Yamaoka Y. Role of vonoprazan in Helicobacter pylori eradication therapy in Japan. Front Pharmacol. 2019; 9:1560. PMID: 30697158.
156. Lee JW, Kim N, Nam RH, Yu JE, Son JH, Lee SM, et al. Efficacy of tegoprazan for improving the susceptibility of antimicrobial agents against antibiotic-resistant Helicobacter pylori . Gut Liver. 2021; 15(1):53–60. PMID: 33191308.
157. Yeom DH, Kim YS. History and pharmacological mechanism of gastric acid-suppressive drugs. Korean J Helicobacter Up Gastrointest Res. 2023; 23(3):159–166.
158. Murakami K, Sakurai Y, Shiino M, Funao N, Nishimura A, Asaka M. Vonoprazan, a novel potassium-competitive acid blocker, as a component of first-line and second-line triple therapy for Helicobacter pylori eradication: a phase III, randomised, double-blind study. Gut. 2016; 65(9):1439–1446. PMID: 26935876.
159. Chey WD, Mégraud F, Laine L, López LJ, Hunt BJ, Howden CW. Vonoprazan triple and dual therapy for Helicobacter pylori infection in the United States and Europe: randomized clinical trial. Gastroenterology. 2022; 163(3):608–619. PMID: 35679950.
160. Yang C, Li S, Huang T, Lin H, Jiang Z, He Y, et al. Effectiveness and safety of vonoprazan-based regimen for Helicobacter pylori eradication: a meta-analysis of randomized clinical trials. J Clin Pharm Ther. 2022; 47(7):897–904. PMID: 35247003.
161. Kato M, Ota H, Okuda M, et al. Guidelines for the management of Helicobacter pylori infection in Japan: 2016 Revised Edition. Helicobacter. 2019; 24(4):e12597. PMID: 31111585.
162. Ouyang Y, Wang M, Xu YL, Zhu Y, Lu NH, Hu Y. Amoxicillin-vonoprazan dual therapy for Helicobacter pylori eradication: a systematic review and meta-analysis. J Gastroenterol Hepatol. 2022; 37(9):1666–1672. PMID: 35716370.
163. Suzuki S, Kusano C, Horii T, Ichijima R, Ikehara H. The ideal Helicobacter pylori treatment for the present and the future. Digestion. 2022; 103(1):62–68. PMID: 34662879.
164. Graham DY. Why the vonoprazan Helicobacter pylori therapies in the US-European trial produced unacceptable cure rates. Dig Dis Sci. 2023; 68(5):1691–1697. PMID: 36856926.
165. Sue S, Maeda S. Is a potassium-competitive acid blocker truly superior to proton pump inhibitors in terms of Helicobacter pylori eradication? Gut Liver. 2021; 15(6):799–810. PMID: 33850058.
166. Mohtasham M, Joukar F, Maroufizadeh S, Mojtahedi K, Asgharnezhad M, Mansour-Ghanaei F. Lactobacillus ruteri compared with placebo as an adjuvant in quadruple therapy for Helicobacter pylori eradication: a randomized, double-blind, controlled trial. Arab J Gastroenterol. 2023; 24(1):40–44. PMID: 36372695.
167. Wang Y, Wang X, Cao XY, Zhu HL, Miao L. Comparative effectiveness of different probiotics supplements for triple Helicobacter pylori eradication: a network meta-analysis. Front Cell Infect Microbiol. 2023; 13:1120789. PMID: 37256113.
168. Hsieh PS, Tsai YC, Chen YC, Teh SF, Ou CM, King VA. Eradication of Helicobacter pylori infection by the probiotic strains Lactobacillus johnsonii MH-68 and L. salivarius ssp. salicinius AP-32. Helicobacter. 2012; 17(6):466–477. PMID: 23067294.
169. Gotteland M, Brunser O, Cruchet S. Systematic review: are probiotics useful in controlling gastric colonization by Helicobacter pylori? Aliment Pharmacol Ther. 2006; 23(8):1077–1086. PMID: 16611267.
170. Oh B, Kim BS, Kim JW, Kim JS, Koh SJ, Kim BG, et al. The effect of probiotics on gut microbiota during the Helicobacter pylori eradication: randomized controlled trial. Helicobacter. 2016; 21(3):165–174. PMID: 26395781.
171. Yuan Z, Xiao S, Li S, Suo B, Wang Y, Meng L, et al. The impact of Helicobacter pylori infection, eradication therapy, and probiotics intervention on gastric microbiota in young adults. Helicobacter. 2021; 26(6):e12848. PMID: 34448282.
172. Huynh HQ, Couper RT, Tran CD, Moore L, Kelso R, Butler RN. N-acetylcysteine, a novel treatment for Helicobacter pylori infection. Dig Dis Sci. 2004; 49(11-12):1853–1861. PMID: 15628716.
173. Gurbuz AK, Ozel AM, Ozturk R, Yildirim S, Yazgan Y, Demirturk L. Effect of N-acetyl cysteine on Helicobacter pylori . South Med J. 2005; 98(11):1095–1097. PMID: 16351030.
174. Cammarota G, Branca G, Ardito F, Sanguinetti M, Ianiro G, Cianci R, et al. Biofilm demolition and antibiotic treatment to eradicate resistant Helicobacter pylori: a clinical trial. Clin Gastroenterol Hepatol. 2010; 8(9):817–820.e3. PMID: 20478402.
175. Chen X, Li P, Shen Y, Zou Y, Yuan G, Hu H. Rhamnolipid-involved antibiotics combinations improve the eradication of Helicobacter pylori biofilm in vitro: a comparison with conventional triple therapy. Microb Pathog. 2019; 131:112–119. PMID: 30951818.
176. Brüssow H. Infection therapy: the problem of drug resistance - and possible solutions. Microb Biotechnol. 2017; 10(5):1041–1046. PMID: 28737021.
177. Loc-Carrillo C, Abedon ST. Pros and cons of phage therapy. Bacteriophage. 2011; 1(2):111–114. PMID: 22334867.
178. Vale FF, Nunes A, Oleastro M, Gomes JP, Sampaio DA, Rocha R, et al. Genomic structure and insertion sites of Helicobacter pylori prophages from various geographical origins. Sci Rep. 2017; 7(1):42471. PMID: 28205536.
179. Lehours P, Vale FF, Bjursell MK, Melefors O, Advani R, Glavas S, et al. Genome sequencing reveals a phage in Helicobacter pylori . mBio. 2011; 2(6):e00239-11. PMID: 22086490.
180. Abdel-Haliem ME, Askora A. Isolation and characterization of bacteriophages of Helicobacter pylori isolated from Egypt. Future Virol. 2013; 8(8):821–826.
181. Cuomo P, Papaianni M, Fulgione A, Guerra F, Capparelli R, Medaglia C. An innovative approach to control H. pylori-induced persistent inflammation and colonization. Microorganisms. 2020; 8(8):1214. PMID: 32785064.
182. Michetti P, Kreiss C, Kotloff KL, Porta N, Blanco JL, Bachmann D, et al. Oral immunization with urease and Escherichia coli heat-labile enterotoxin is safe and immunogenic in Helicobacter pylori-infected adults. Gastroenterology. 1999; 116(4):804–812. PMID: 10092302.
183. Kreiss C, Buclin T, Cosma M, Corthésy-Theulaz I, Michetti P. Safety of oral immunisation with recombinant urease in patients with Helicobacter pylori infection. Lancet. 1996; 347(9015):1630–1631.
184. DiPetrillo MD, Tibbetts T, Kleanthous H, Killeen KP, Hohmann EL. Safety and immunogenicity of phoP/phoQ-deleted Salmonella typhi expressing Helicobacter pylori urease in adult volunteers. Vaccine. 1999; 18(5-6):449–459. PMID: 10519934.
185. Angelakopoulos H, Hohmann EL. Pilot study of phoP/phoQ-deleted Salmonella enterica serovar typhimurium expressing Helicobacter pylori urease in adult volunteers. Infect Immun. 2000; 68(4):2135–2141. PMID: 10722611.
186. Kotloff KL, Sztein MB, Wasserman SS, Losonsky GA, DiLorenzo SC, Walker RI. Safety and immunogenicity of oral inactivated whole-cell Helicobacter pylori vaccine with adjuvant among volunteers with or without subclinical infection. Infect Immun. 2001; 69(6):3581–3590. PMID: 11349017.
187. Bumann D, Metzger WG, Mansouri E, Palme O, Wendland M, Hurwitz R, et al. Safety and immunogenicity of live recombinant Salmonella enterica serovar Typhi Ty21a expressing urease A and B from Helicobacter pylori in human volunteers. Vaccine. 2001; 20(5-6):845–852. PMID: 11738748.
188. Sougioultzis S, Lee CK, Alsahli M, Banerjee S, Cadoz M, Schrader R, et al. Safety and efficacy of E. coli enterotoxin adjuvant for urease-based rectal immunization against Helicobacter pylori . Vaccine. 2002; 21(3-4):194–201. PMID: 12450694.
189. Banerjee S, Medina-Fatimi A, Nichols R, Tendler D, Michetti M, Simon J, et al. Safety and efficacy of low dose Escherichia coli enterotoxin adjuvant for urease based oral immunisation against Helicobacter pylori in healthy volunteers. Gut. 2002; 51(5):634–640. PMID: 12377799.
190. Metzger WG, Mansouri E, Kronawitter M, Diescher S, Soerensen M, Hurwitz R, et al. Impact of vector-priming on the immunogenicity of a live recombinant Salmonella enterica serovar typhi Ty21a vaccine expressing urease A and B from Helicobacter pylori in human volunteers. Vaccine. 2004; 22(17-18):2273–2277. PMID: 15149786.
191. Aebischer T, Bumann D, Epple HJ, Metzger W, Schneider T, Cherepnev G, et al. Correlation of T cell response and bacterial clearance in human volunteers challenged with Helicobacter pylori revealed by randomised controlled vaccination with Ty21a-based Salmonella vaccines. Gut. 2008; 57(8):1065–1072. PMID: 18417532.
192. Malfertheiner P, Selgrad M, Wex T, Romi B, Borgogni E, Spensieri F, et al. Efficacy, immunogenicity, and safety of a parenteral vaccine against Helicobacter pylori in healthy volunteers challenged with a Cag-positive strain: a randomised, placebo-controlled phase 1/2 study. Lancet Gastroenterol Hepatol. 2018; 3(10):698–707. PMID: 30042064.
193. Malfertheiner P, Schultze V, Rosenkranz B, Kaufmann SH, Ulrichs T, Novicki D, et al. Safety and immunogenicity of an intramuscular Helicobacter pylori vaccine in noninfected volunteers: a phase I study. Gastroenterology. 2008; 135(3):787–795. PMID: 18619971.
194. Zeng M, Mao XH, Li JX, Tong WD, Wang B, Zhang YJ, et al. Efficacy, safety, and immunogenicity of an oral recombinant Helicobacter pylori vaccine in children in China: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2015; 386(10002):1457–1464. PMID: 26142048.
TOOLS
Similar articles