Open access peer-reviewed chapter

Helicobacter pylori Challenge Vaccine for Humans

Written By

Rike Syahniar, Dayu Swasti Kharisma and Rayhana

Submitted: 16 September 2021 Reviewed: 10 October 2021 Published: 26 November 2021

DOI: 10.5772/intechopen.101157

From the Edited Volume

Vaccine Development

Edited by Yulia Desheva

Chapter metrics overview

450 Chapter Downloads

View Full Metrics

Abstract

Helicobacter pylori infect during childhood and are typically present for life, despite a vigorous host immune response, which includes the invading pathogen being coated with antibodies. This bacterial longevity indicates the development, on the part of the pathogen, of a range of processes for evading effective host immunity. Since its discovery 25 years ago, significant progress has been made in understanding the virulence factors and several aspects of the pathogenesis of H. pylori gastric diseases. The prevalence of antimicrobial drug resistance is so high that all patients infected with H. pylori should be considered resistant infections. The most severe consequence of H. pylori infection, and the key reason a vaccine is required, is gastric cancer, globally the third leading cause of death due to cancer. Patients typically present with gastric cancer without knowing they are infected; eradication likely has little effect by this time. Vaccine against H. pylori that reduces the incidence of gastric cancer will probably be cost effective in developed countries. Several vaccines were successfully tested in different experimental animal models, but translation into an efficacious human vaccine has been unsuccessful.

Keywords

  • Helicobacter pylori
  • H. pylori vaccine
  • gastric disease
  • gastric cancer

1. Introduction

Helicobacter pylori (H. pylori) is a gram-negative, motile, spiral, or curved bacterium that colonizes the human gastric mucosa about 50% of the human population [1]. H. pylori induces the development of a peptic ulcer, gastric mucosa-associated lymphoid tissue lymphoma, and gastric cancer [2]. Globally, gastric cancer ranks as the third leading cause of death from malignancy [3].

The prevalence of H. pylori infection in developed countries ranges from 30 to 40%, while it can reach 80% [4, 5, 6, 7, 8]. Hygiene conditions and socioeconomic status affect the incidence of H. pylori infection [5]. H. pylori infection is thought to result from direct human-to-human transmission through oral, fecal, or both. The infection is generally acquired during childhood and then increases gradually with age. H. pylori is commonly transmitted from mother to child [1] This evidence is supported by the ability to grow H. pylori from vomit or oral area and analysis of bacterial strains indicating general vertical transmission between mothers and their offspring [1, 9, 10]. If left untreated, most H. pylori infections last a lifetime.

It is challenging to eradicate H. pylori because of its high antimicrobial resistance. In addition, most of the infected people are asymptomatic. The costs for adequate diagnostic tests and pharmacological eradication will be enormous. Treatment of H. pylori requires a multidrug regimen because the organism colonizes beneath the mucous layer, which reduces the direct effect of antibiotic penetration [11]. Resistance is also a problem with some commonly used antibiotics, namely metronidazole, amoxicillin, erythromycin, and clarithromycin [11, 12, 13, 14, 15]. Unfortunately, increasing antibiotic resistance is beginning to affect the efficacy of treatment, and, despite the impact of H. pylori, preventive vaccination strategies are still lacking. Until now, there is no recommended H. pylori vaccine available. Here, we provide an overview of the constraints and challenges in the manufacture of vaccines against H. pylori.

Advertisement

2. Morphology of H. pylori

H. pylori is a gram-negative, non-spore-forming bacterium, spiral-shaped or rods. It will turn into a coccoid form in unfavorable conditions, a form of defense resistant to conditions [16]. Several studies have shown that the coccoid form of H. pylori decreases morphological manifestations and remains alive and metabolizes actively, although it cannot be cultured [16, 17].

These bacteria have flagella that allow high motility and are microaerophilic. H. pylori has 1–6 sheathed flagella at the terminals, which are lophotrically arranged. Other Helicobacter species have flagella that are not sheathed [18]. The length of this bacterium is between 2.5 and 3.5 m, and the width is 0.5–1.0 m. H. pylori can grow well at 35–37°C and produces the enzyme catalase, cytochrome oxidase, urease, alkaline phosphatase, and glutamyl transpeptidase. The proper place for bacteria to live in the human body is the antrum. The number of H. pylori that appears to show the ability to adapt to certain areas, for example, in the human stomach on the surface of epithelial cells and the mucous layer [16, 18].

These bacteria have the same layer composition as other gram-negative bacteria: an inner membrane, periplasm with peptidoglycan, and an outer membrane. The outer membrane is composed of phospholipids and lipopolysaccharides (LPS). H. pylori LPS variation plays a role in population heterogeneity and allows adaptation to changes in gastric mucosal conditions. Outer membrane phospholipids contain cholesterol glucoside, which is very rare in bacteria [18].

Advertisement

3. H. pylori adaptation and colonization

After entering the stomach, H. pylori performs four colonizing stages, including surviving against gastric acid, moving toward gastric epithelial cells, attaching to gastric epithelial cells, and releasing toxins, causing tissue damage [19]. The enzyme urease is an essential factor for H. pylori colonization of the gastric mucosa. This enzyme converts urea (a product secreted by gastric cells) into ammonia and carbon dioxide. Ammonia can increase the pH of the gastric mucosa around bacterial cells. Therefore, this enzyme neutralizes the acidity of the stomach and aids in colonization. However, this enzyme can also stimulate monocytes, neutrophil chemotaxis, and stimulate cytokine production [20].

H. pylori uses flagella and specific chemoreceptors, TIpB, to move toward gastric epithelial cells near-neutral pH. The circular motion is facilitated by the helical shape of the bacteria, which can pass through the dense mucosal layer [20]. These bacteria can penetrate the mucus layer through the production of a protein called collagenase/mucinase. Collagenase/mucinase functions to liquefy mucin, reducing viscosity and allowing these bacteria to move more freely to reach epithelial cells. In addition, H. pylori produces alpha-carbonic anhydrase (α-CA), which helps urease convert carbon dioxide into bicarbonate. Bicarbonate is a weak base that can neutralize stomach acidity [19].

After successfully passing gastric acid, H. pylori attaches to gastric epithelial cells with the help of adhesins. Adhesins bind to receptors on the gastric mucosal surface. The majority of adhesins are H. pylori outer membrane proteins (OMPs) [21]. These adhesives include BabA (Blood Group Antigen-Binding Adhesin), SabA (Sialic acid-binding Adhesin), AlpA/B (Adherence-associated Lipoproteins A/B), HopZ, OipA, and HpaA. Receptors for BabA, Sab A, and AlpA/B adhesins include the Lewis human blood antigen group b (Leb), sialyl Lex, and laminin. Meanwhile, the HopZ and OipA receptors have not been identified [21, 22, 23].

The LPS chemical structure of several H. pylori strains resembles the Lewis x and Lewis y blood antigen groups expressed in the gastric mucosa. It serves to downregulate the immune response in patients with acute and chronic infections [24]. Specific modification of the LPS molecule allows molecular mimicry and alteration of the structural components of lipid A, leading to low endotoxic activity. The H. pylori membrane is coated with the same molecules on the host cell as plasminogen and cholesterol that protect the bacteria from host cell recognition. The high genetic diversity of bacteria allows for rapid adaptation to environmental changes [20]. Phospholipase H. pylori produces products such as lysolecithin, which interfere with the protective layer of phospholipids that are abundant in the apical membrane of mucus cells [24].

H. pylori has a Cag Pathogenicity Island (CagPAI), which is associated with the development of chronic active gastritis, peptic ulcer, and atrophic gastritis with an increased risk of gastric cancer. CagA is a virulence factor located at one end of CagPAI, a 40 kb of the H. pylori genome. Cag PAI encodes 31 genes that make up the type IV secretory system (T4SS), which injects CagA, an oncoprotein, into the cytosol of gastric epithelial cells [23]. Upon entry into gastric epithelial cells, CagA undergoes Src-dependent tyrosine phosphorylation and activates SHP-2, which leads to dephosphorylation of host cell proteins and changes in cellular morphology. Translocation of CagA protein supports the release of essential nutrients to the apical side of epithelial cells, either by induction of inflammation or by host cell depolarization [24]. Apart from CagA, peptidoglycan is also transported into the host cell via T4SS and outer membrane vesicles. Peptidoglycan activates the intracellular Nod1 receptor, which activates NF-kB (a transcription factor associated with epithelial gene expression and regulates the expression of various proinflammatory cytokines). Modifications in these settings are important to dampen the host immune system and contribute to bacterial persistence [25].

The second most studied H. pylori virulence factor is VacA. VacA is a 140-kDa polypeptide. The gene-encoding VacA is present in all H. pylori strains and exhibits allelic diversity in three major regions, namely, s (signal), the i (intermediate), and m (middle). As a result, the cytotoxic activity of the toxin varies between strains. Different combinations of alleles from each region (s1, s2, i1, i2, m1, m2) that exist result in different abilities of VacA toxin to stimulate vacuolation in epithelial cells [24]. VacA can bind to several epithelial cell surface molecules, including transmembrane protein receptors-type tyrosine-protein phosphatase (PTPRZ1), fibronectin, EGFR, CD18 on T-cells, and various lipids and sphingomyelin [24]. VacA can induce vacuolation and several cellular activities, including membrane channel formation, the release of cytochrome c from mitochondria leading to apoptosis, and binding to cell membrane receptors followed by initiation of the proinflammatory response. VacA can also inhibit the activation and proliferation of T and B cells [23]. VacA can also inhibit the phagosomal maturation of macrophages and induce macrophage apoptosis [24].

Advertisement

4. Natural history of H. pylori infection

The natural history of H. pylori infection can be divided into two stages. The first is the acute phase, where bacteria multiply and cause gastric inflammation, hypochlorhydria, and some gastrointestinal symptoms such as fullness in the stomach, nausea, and vomiting [26]. This phase often occurs during childhood and is almost difficult to diagnose. After several weeks, the chronic phase begins with a reduced inflammatory response, and gastric pH normalizes, then becomes asymptomatic. Colonization of H. pylori in the gastric mucosa causes infiltration of neutrophils and mononuclear cells in the antrum and body, leading to chronic inflammation. When colonization becomes persistent, there is a close correlation between the level of acid secretion and the distribution of chronic gastritis. The most common feature is non-atrophic gastritis with normal acid secretion in asymptomatic subjects. Antral-dominant gastritis is associated with hyperchlorhydria and duodenal ulcers, whereas dominant corpus gastritis causes hypochlorhydria, gastric atrophy, intestinal metaplasia, and an increased risk of distal gastric cancer [27].

H. pylori colonization causes a degree of inflammation in the gastric mucosa. Reactive oxygen species produced from polymorphonuclear after activation by H. pylori induce gastric mucosal injury. Polymorphonuclear cell infiltration of the gastric mucosa leads to the development of early H. pylori infection lesions called active chronic gastritis, the natural history of H. pylori infection [16].

Acid secretion is affected by H. pylori infection, which is also associated with dyspepsia. Patients with functional dyspepsia and H. pylori infection had a fourfold increase in acid secretion. In contrast, asymptomatic H. pylori-positive individuals had only a 2.5-fold increase in acid secretion. Acid secretion during H. pylori infection depends on the spread of gastric mucosal atrophy and the local stage of inflammation, which is determined by interactions between the host, bacteria, and environmental factors. In H. pylori-infected patients with dominant antral gastritis without corpus atrophy, acid secretion was more elevated than in uninfected normal mucosa. This is a potential cause of dyspeptic symptoms, such as epigastric pain or burning. In contrast, when the atrophy extends to the corpus mucosa (fundic glands), reduced acid secretion is due to direct damage to parietal cells in the corpus, associated with gastric ulcers and gastric cancer [16, 28, 29].

Advertisement

5. Immune evasion in persistent infection with H. pylori

The primary defense barrier against H. pylori is the mucus secreted by epithelial cells and innate immune cells in the lamina propria [30]. H. pylori and its products can directly contact lamina propria immune cells, resulting in an influx of immune cells that include neutrophils, macrophages, dendritic cells, lymphocytes, and associated innate and adaptive immune responses [31]. Toll-like receptors (TLRs) are a major group of Pattern Recognition Receptors (PRRs) that recognize pathogen-associated molecular patterns (PAMPs). Bacterial lipopolysaccharide (LPS), peptidoglycan, lipoprotein, lipoteichoic acid, and unmethylated CpG-rich regions of DNA are the main targets of TLRs [31]. Studies have shown that H. pylori has managed to escape the introduction of TLRs. For example, TLR4 runs the well-described LPS recognition [32].

H. pylori subverts the adaptive immune response by modulating effector T cells [31]. During H. pylori infection, the frequency of CD4+ T cells in the gastric lamina propria with a memory phenotype increases and polarizes to a Th1/Th17 phenotype, but these T cells are hyporesponsive to this bacterium [33]. Because this hyporesponsiveness contributes to chronic infection, attempts have been made by H. pylori to downregulate the T-cell response. H. pylori also manipulate T cell function by eliciting regulatory T cells (Tregs), frequently found in these patients [34]. Unusual Tregs activation by microbial antigens may provide a mechanism for preventing H. pylori from the immune response. Gamma-glutamyl transpeptidase (GGT) and VacA from the H. pylori molecule indirectly affect T lymphocyte activity and promote the differentiation of effector CD4+ T cells into Tregs. [35].

Advertisement

6. Current status and challenges of H. pylori vaccine candidate development

Since the early 1990s, vaccines based on various antigens, adjuvants, and routes of administration have been evaluated. The mucosal route of administration, especially the oral route, is the most suitable route for vaccination against H. pylori infection to induce an effective immune response at the site of infection. Until now, many vaccine candidates have been developed at the preclinical stage. In comparison, the most advanced candidate for the H. pylori vaccine is in phase 3 clinical trials (Table 1).

Candidate vaccineCountry of the laboratoryTrial statusProphylactic /therapeuticReferences
Candidate vaccine: Oral Recombinant Helicobacter Pylori VaccineChinaPhase IIIProphylactic[36]
Imevax/IMX101GermanyPhase IProphylactic[37]
HelicoVaxUSAPreclinicalTherapeutic[38]
Recombinant CTB-UreI-UreB (BIB)ChinaPreclinicalProphylactic[39]
Recombinant Vibrio cholerae expressing H. pylori HpaA antigenSwedenPreclinicalProphylactic[40]
CTB-Lpp20ChinaPreclinicalProphylactic /Therapeutic[41]
MCRI (Murdoch Children Research Institute /Gastric Cancer Vaccines)AustraliaPreclinicalProphylactic[42]
H. pylori Vaccine (NCT00736476)GermanyI/IIProphylactic[43]

Table 1.

Summary of H. pylori vaccine development status.

6.1 Oral recombinant Helicobacter pylori vaccine

This vaccine (UreB/LTB fusion vaccine) is a recombinant oral H. pylori vaccine using a urease B subunit (gene derived from H. pylori 9803) fused with heat-labile enterotoxin B subunit (gene derived from E.coli H44815) developed by Third Military Medical University and Chongqing KangWei Biotechnology in China. A randomized, double-blind, placebo-controlled phase III clinical trial was conducted in Ganyu County, Jiangsu Province, China. Vaccination was administered orally in 3 doses on days 0, 14, and 28. In this study, oral administration of the H. pylori vaccine provided good protection against H. pylori infection in children aged 6–15 years up to 1 year after vaccination. Although the estimated point of protection for the vaccine later shows a slight decrease in efficacy, overall safety can last up to 3 years. All side effects are mild and improve within 24 hours. The most common reaction is vomiting, followed by fever and headache [36].

6.2 Imevax/IMX101

Imevax has completed a phase I clinical trial with IMX101. The vaccine consists of the H. pylori antigen-glutamyltranspeptidase (GGT), an outer membrane protein, and a mucosal adjuvant. The main reason for the failure of previous vaccines to provide complete protection is the immune evasion strategy possessed by H. pylori [35].

One of the most important is GGT, which appears to have relatively immunosuppressive solid activity. Therefore, this vaccine aims to target and neutralize these defense mechanisms, potentially enabling a more effective protective immune response against other antigenic components of the vaccine. The phase I clinical trial of Imevax IMX101 was conducted in a multi-center, randomized, double-blind, and adjuvant-controlled study conducted on adult volunteers aged 18 to 50 years to evaluate safety, tolerability, and efficacy. Volunteers consisted of negative people for H. pylori and healthy people infected with H. pylori. IMX101 vaccine is administered sublingually and intradermally [37].

6.3 HelicoVax

In the study by Steven F. Moss et al., they designed two DNA vaccines, namely HelicoVax A and HelicoVaxB, each containing a set of 25 different HLA class II epitopes. Steven F. Moss et al. used C57BL/6 mice. At six weeks, mice were infected with H. pylori strain SS1 in 0.1 ml PBS, 3 times in 1 week. DNA vaccines are administered intramuscularly and intranasally. The test results show that there is promising therapeutic efficacy for the development of an epitope-based mucosal vaccine against H. pylori.

6.4 Recombinant CTB-UreI-UreB (BIB)

Epitope vaccines are a promising option for protection against H. pylori infection. Research conducted by Jing Yang et al. developed a multi-epitope vaccine by linking the cholera toxin B (CTB) subunit, two antigenic fragments of H. pylori urease I subunit (UreI20–29, UreI98–107), and 4 H. pylori antigenic fragments. Urease B subunit, (UreB12–23,UreB229–251,UreB327–400,UreB515–561) produces recombinant CTB-Urel-UreB (BIB). This vaccine’s protective effect against H. pylori infection was evaluated in BALB/c mice. Significant protection against H. pylori infection was achieved in BALB/c mice immunized with BIB, rIB plus rCTB, and rIB. Induction of substantial protection against H. pylori was mediated by specific serum IgA and mucosal sIgA antibodies and a mixed response of Th1/Th2/Th17 cells. This multi-epitope vaccine can be a promising vaccine candidate that helps control H. pylori infection [39].

6.5 Recombinant Vibrio cholerae expressing H. pylori HpaA antigen

The vaccine was designed by constructing and characterizing the faster-growing O1 Vibrio cholerae strain of H. pylori as a vector for the H. pylori antigen that might be used as a vaccine strain against H. pylori. Joshua Tobias et al. developed the technology of over-expressing enterotoxigenic E. coli (ETEC) colonization factor antigens (CFs), the main virulence factor of ETEC, in heterologous bacterial strains including V.cholerae. Due to the extracellular nature of H. pylori, the bacteria colonize the epithelial surface and coat the gastric mucosal lining and areas of gastric metaplasia in the duodenum. Joshua Tobias et al. constructed a V.cholerae strain that overexpressed HpaA, as a surface antigen and H. pylori-specific lipoprotein known to mediate H. pylori colonization in the rat stomach and be a protective antigen against H. pylori infection in animal models, singly or in animal models. Concurrently with different ETEC CFs can promote bacterial binding to the small intestinal mucosa. Specific strains were developed and characterized to the level of surface expression of HpaA, and the capacity to induce an immune response against H. pylori in mice after oral immunization [40].

6.6 Lp220 vaccine

Epitope vaccine is a potential vaccine as a prophylactic and therapeutic vaccine against H. pylori infection. Lpp20 is one of the main protective antigens that trigger immune responses after H. pylori invades the host and is considered an excellent vaccine candidate for the control of H. pylori infection. This epitope vaccine consists of a mucosal adjuvant cholera toxin B subunit (CTB) and three Lpp20 epitopes that have been identified (one B cell epitope and two CD4+ T cell epitopes) for efficacy in mice. An epitope vaccine consisting of CTB, one B cell, and 2 CD4+ T cell epitopes of Lpp20 was prepared and named CTB-Lpp20, which is expressed in Escherichia coli and used for immunization BALB/c mice via intragastric. The CTB-Lpp20 epitope vaccine has good immunogenicity and immunoreactivity and can produce high specific antibodies against Lpp20 and the cytokines IFN-c and IL-17. In addition, CTB-Lpp20 significantly decreased H. pylori colonization in mice. This protection correlates with IgG, IgA, and sIgA antibodies and Th1-type cytokines [41].

6.7 MCRI (Murdoch children research institute/gastric cvaccines)

MCRI (Murdoch Children Research Institute) developed a new vaccine strategy that prevents H. pylori-induced inflammation. The vaccine candidates are recombinant HtrA, a-55 kDa protein, and the only serine protease produced by H. pylori. HtrA is expressed and secreted on the bacterial surface, and is essential for the survival of H. pylori. MCRI investigators have shown that mice vaccinated with HtrA protected against H. pylori-induced inflammation compared with controls. H. pylori HtrA destroys the epithelial barrier by cleaving E-cadherin thereby opening junctions between gastric epithelial cells. The leaky epithelium allows a number of bacteria to enter the epithelial barrier to the tissue, interact with immune system cells, and cause gastritis. MCRI investigators found that serum from mice vaccinated with HtrA neutralized HtrA protease activity in vitro [42].

6.8 H. pylori vaccine (NCT00736476)

The vaccine consists of three recombinant H. pylori antigens vacuolating cytotoxin A (VacA), cytotoxin-associated gene A (CagA), and neutrophil-activating protein (NAP) that prevent infection in animal models and are well tolerated and highly immunogenic in adults receiving healthy. In this phase 1/2 randomized, single-center, unsupervised, placebo-controlled study, healthy nonpregnant adults aged 18–40 years who were confirmed negative for H. pylori infection were randomly assigned (3:4) to three intramuscular doses placebo or vaccine at 0, 1, and 2 months [43]. Previously, three recombinant antigen vaccines were tested relevant to H pylori virulence—CagA, VacA, and NAP—in phase 1 clinical study [44]. Compared with placebo, the vaccine did not provide additional protection against H. pylori infection after challenge with CagA-positive strains, despite an increased systemic humoral response to key H. pylori antigens. The vaccine induces high-titer IgG antibodies specific for CagA, VacA, and NAP and a robust antigen-specific T cell response, but this is not sufficient to eliminate H. pylori [43].

Advertisement

7. Conclusion

The best preclinical results are obtained from vaccines that often induce a T-cell-mediated immune response rather than humoral immunity. Th1 and Th17 responses in the stomach are more protective. The mechanisms of H. pylori persistence and the utilization of multiple mechanisms to overcome adaptive immunity are recognized as essential barriers to vaccination. Several vaccines were successfully tested in different experimental animal models, but translation into an efficacious human vaccine was unsuccessful. A better understanding of the immune response generated by natural H. pylori infection and the mechanism by which the bacteria survives is needed for the development of human vaccines. Future vaccines for the prevention of H. pylori infection should be conducted in children, where infection occurs naturally. Therefore, prophylactic vaccines may need to be given to children in the first few years of life to reach the maximum number of target groups when uninfected, but the health benefits will emerge five decades later.

Advertisement

Acknowledgments

We would like to thank the Faculty of Medicine and Health, Universitas Muhammadiyah Jakarta for support of this work.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Syahniar R, Mardiastuti, Syam AF, Yasmon A. Phylogenetic analysis of Helicobacter pylori 16S rRNA gene in gastric biopsy from patients with dyspepsia. Advanced Science Letters. 2018;24(9):6789-6792
  2. 2. Tsay F, Hsu P. H. pylori infection and extra-gastroduodenal diseases. Journal of Biomedical Science. 2018;25(65):1-8
  3. 3. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians. 2018;68(6):394-424
  4. 4. JKY H, Ying Lai W, Khoon Ng W, MMY S, Underwood FE, Tanyingoh D, et al. Global prevalence of Helicobacter pylori infection: Systematic review and meta-analysis. 2017 Aug;153(2):420-429 [cited 12 September 2021]; Available from:. DOI: 10.1053/j.gastro.2017.04.022
  5. 5. Wang W, Jiang W, Zhu S, Sun X, Li P, Liu K, et al. Assessment of prevalence and risk factors of helicobacter pylori infection in an oilfield Community in Hebei, China. BMC Gastroenterology. 2019;19(1):4-11
  6. 6. Syam AF, Miftahussurur M, Makmun D, Nusi IA, Zain LH, Zulkhairi, et al. Risk factors and prevalence of Helicobacter pylori in five largest Islands of Indonesia: A preliminary study. PLoS One. 2015 [cited 12 September 2021];10(11):e0140186 Available from: https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0140186
  7. 7. Syahniar R, Wahid MH, Syam AF, Yasmon A. Detecting the Helicobacter pylori 16S rRNA gene in dyspepsia patients using real-time PCR. Acta Medica Indonesiana. 2019;51(1):34-41
  8. 8. Khoder G, Sualeh Muhammad J, Mahmoud I, Soliman SSM, Burucoa C. Prevalence of helicobacter pylori and its associated factors among healthy asymptomatic residents in the united arab emirates. Pathogens. 2019 Apr 1;8(2):44
  9. 9. Konno M, Yokota S, Suga T, Takahashi M, Sato K, Fujii N. Predominance of mother-to-child transmission of Helicobacter pylori infection detected by random amplified polymorphic DNA fingerprinting analysis in Japanese families. The Pediatric Infectious Disease Journal. 2008 [cited: 11 September 2021];27(11):999-1003 Available from: https://pubmed.ncbi.nlm.nih.gov/18845980/
  10. 10. Wahid MH, Yuliantiningsih L, Syahniar R, Syam AF, Yasmon A, Krisnuhoni E, et al. Helicobacter pylori infection in siblings: A case report. In: Yunir E, editor. Medical Case Report. New York: Nova Science Publishers Inc; 2020. pp. 227-232
  11. 11. Kipritci Z, Gurol Y, Celik G. Antibiotic resistance results of Helicobacter pylori in a University Hospital: Comparison of the hybridization test and real-time polymerase chain reaction. International Journal of Microbiology. 2020;2020:1-5
  12. 12. Park JY, Shin TS, Kim JH, Yoon HJ, Kim BJ, Kim JG. The prevalence of multidrug resistance of helicobacter pylori and its impact on eradication in Korea from 2017 to 2019: A single-center study. Antibiotics. 2020;9(10):1-11
  13. 13. Wang D, Guo Q , Yuan Y, Gong Y. The antibiotic resistance of Helicobacter pylori to five antibiotics and influencing factors in an area of China with a high risk of gastric cancer. BMC Microbiology. 2019[cited: 12 September 2021];19(1):1-10 Available from: https://bmcmicrobiol.biomedcentral.com/articles/10.1186/s12866-019-1517-4
  14. 14. Schubert JP, Gehlert J, Rayner CK, Roberts-Thomson IC, Costello S, Mangoni AA, et al. Antibiotic resistance of Helicobacter pylori in Australia and New Zealand: A systematic review and meta-analysis. Journal of Gastroenterology and Hepatology [cited 12 September 2021];2021;36(6):1450-1456 Available from: https://onlinelibrary.wiley.com/doi/full/10.1111/jgh.15352
  15. 15. Li J, Deng J, Wang Z, Li H, Wan C. Antibiotic resistance of Helicobacter pylori strains isolated from pediatric patients in Southwest China. Frontiers in Microbiology. 2021;0:3647
  16. 16. JG K, AH v V, EJ K. Pathogenesis of Helicobacter pylori infection. Clinical Microbiology Reviews. 2006[cited 12 September 2021];19(3):449-490 Available from: https://pubmed.ncbi.nlm.nih.gov/16847081/
  17. 17. Ierardi E, Losurdo G, Mileti A, Paolillo R, Giorgio F, Principi M, et al. The puzzle of coccoid forms of Helicobacter pylori: Beyond basic science. Antibiotics. 2020 [cited 12 September 2021];9(6):1-14 Available from: /pmc/articles/PMC7345126/
  18. 18. Phe. UK Standards for Microbiology Investigations-standards-for-microbiology-investigations-smi-quality-and-consistency-in-clinical-laboratories PHE Publications gateway number: 2015075 UK Standards for Microbiology Investigations are produced in association with. 2015
  19. 19. Kao CY, Sheu BS, Wu JJ. Helicobacter pylori infection: An overview of bacterial virulence factors and pathogenesis. Biomedical Journal. 2016 [cited 12 September 2021];39(1):14-23 Available from: https://pubmed.ncbi.nlm.nih.gov/27105595/
  20. 20. Bauer B, Meyer TF. The human gastric pathogen Helicobacter pylori and its association with gastric cancer and ulcer disease. Ulcers. 2011;18(2011):1-23
  21. 21. Kim N. Prevalence and transmission routes of H. pylori. Helicobacter pylori. 2016:3-19
  22. 22. Oleastro M, Ménard A. The role of Helicobacter pylori outer membrane proteins in adherence and pathogenesis. Biology (Basel). 2013 [cited 13 September 2021];2(3):1110 Available from: /pmc/articles/PMC3960876/
  23. 23. Shiota S, Suzuki R, Yamaoka Y. The significance of virulence factors in Helicobacter pylori. J Dig Dis. 2013 [cited 13 September 2021];14(7):341-349 Available from: https://pubmed.ncbi.nlm.nih.gov/23452293/
  24. 24. Roesler BM, Rabelo-Gonçalves EMA, Zeitune JMR. Virulence Factors of Helicobacter pylori: A Review. Clinical Medicine Insights: Gastroenterology. 2014 [cited 13 September 2021];7(7):9 Available from: /pmc/articles/PMC4019226/
  25. 25. Kalali B, Mejías-Luque R, Javaheri A, Gerhard M. H. pylori virulence factors: influence on immune system and pathology. Mediators Inflamm. 2014;2014:426309. doi: 10.1155/2014/426309. Epub 2014 Jan 21. PMID: 24587595; PMCID: PMC3918698
  26. 26. Portal-Celhay C, Perez-Perez GI. Immune responses to Helicobacter pylori colonization: mechanisms and clinical outcomes. Clinical Science (London). 2006 [cited 13 September 2021];110(3):305-314 Available from: https://pubmed.ncbi.nlm.nih.gov/16464172/
  27. 27. Phan Trung N. International PhD School in Biomolecular and Biotechnological Sciences Cycle XXVII Subject: Clinical and Molecular Microbiology. Polymorphisms of cagA and vacA Genes in Helicobacter pyloriIsolated from Gastroduodenal Diseases Patients in Central Vietnam Originality Statement Dissertation: INTERNATIONAL PhD SCHOOL IN Biomolecular and Biotechnological Sciences Cycle XXVII. Vietnam
  28. 28. Suzuki H, Moayyedi P. Helicobacter pylori infection in functional dyspepsia. Nature Reviews Gastroenterology & Hepatology. 2013 [cited 13 September 2021];10(3):168-174 Available from: https://pubmed.ncbi.nlm.nih.gov/23358394/
  29. 29. Kim Y-J, Chung WC, Kim BW, Kim SS, Il KJ, Kim NJ, et al. Is Helicobacter pylori associated functional dyspepsia correlated with dysbiosis? Journal of Neurogastroenterology and Motility. 2017 [cited 13 September 2021];23(4):504-516 Available from: https://www.jnmjournal.org/journal/view.html?doi=10.5056/jnm17066
  30. 30. Chmiela M, Karwowska Z, Gonciarz W, Allushi B, Stączek P. Host pathogen interactions in Helicobacter pylori related gastric cancer. http://www.wjgnet.com/ 2017 [cited 13 September 2021];23(9):1521-1540. Available from: https://www.wjgnet.com/1007-9327/full/v23/i9/1521.htm
  31. 31. Nouri A. Helicobacter pylori evasion strategies of the host innate and adaptive immune responses to survive and develop gastrointestinal diseases | Elsevier Enhanced Reader. Microbiological Research. 2019 [cited 13 Septermber 2021];218:49-57 Available from: https://reader.elsevier.com/reader/sd/pii/S0944501318308358?token=4B14C32FD720DDC898F6AA5065BAD415012D776721C57A5CE5A4414CB111011F16EA478013FEEA57B8759EC1CEC37B61&originRegion=eu-west-1&originCreation=20210913012435
  32. 32. Stead CM, Beasley A, Cotter RJ, Trent MS. Deciphering the unusual acylation pattern of Helicobacter pylori lipid A. Journal of Bacteriology. 2008 [cited 13 September 2021];190(21):7012 Available from: /pmc/articles/PMC2580709/
  33. 33. Lina TT, Alzahrani S, House J, Yamaoka Y, Sharpe AH, Rampy BA, Pinchuk IV, Reyes VE. Helicobacter pylori cag pathogenicity island’s role in B7-H1 induction and immune evasion. PLoS One. 2015; [cited 14 Sepetmber 2021];10(3) Available from: https://pubmed.ncbi.nlm.nih.gov/25807464/
  34. 34. Bagheri N, Shirzad H, Elahi S, Azadegan-Dehkordi F, Rahimian G, Shafigh M, et al. Downregulated regulatory T cell function is associated with increased Peptic Ulcer in Helicobacter pylori-infection. Microbial Pathogenesis. 2017;110:165-175
  35. 35. Oertli M, Noben M, Engler DB, Semper RP, Reuter S, Maxeiner J, et al. Helicobacter pylori γ-glutamyl transpeptidase and vacuolating cytotoxin promote gastric persistence and immune tolerance. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(8):3047-3052
  36. 36. Zeng M, Mao X-H, Li J-X, Tong W-D, Wang B, Zhang Y-J, et al. Articles 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 Oct 10;386(10002):1457-64 [cited 14 Septermber 2021] . DOI: 10.1016/S0140-6736Available from:
  37. 37. Phase 1a/b Study on Safety of IMX101 in H. Pylori-negative and H. Pylori-infected Healthy Volunteers - Full Text View - ClinicalTrials.gov. [cited 14 September 2021]. Available from: https://clinicaltrials.gov/ct2/show/NCT03270800
  38. 38. Moss SF, Moise L, Lee DS, Kim W, Zhang S, Lee J, et al. HelicoVax: Epitope-based therapeutic H. pylori vaccination in a mouse model. Vaccine. 2011;29(11):2085-2091
  39. 39. Wang B, Pan X, Wang H, Zhou Y, Zhu J, Yang J, et al. Immunological response of recombinant H. pylori multi-epitope vaccine with different vaccination strategies. International Journal of Clinical and Experimental Pathology. 2014 [cited 14 september 2021];7(10):6559-6566 Available from: https://europepmc.org/articles/PMC4230088
  40. 40. Tobias J, Lebens M, Sun Nyunt W, Holmgren J, Svennerholm A-M. Surface expression of Helicobacter pylori HpaA adhesion antigen on Vibrio cholerae, enhanced by co-expressed enterotoxigenic Escherichia coli fimbrial antigens. Microbial Pathogenesis. 2017 [cited 15 September 2021];105:177-184 Available from: https://pubmed.ncbi.nlm.nih.gov/28215587/
  41. 41. Li Y, Chen Z, Ye J, Ning L, Luo J, Zhang L, Jiang Y, Xi Y, Ning Y. Antibody production and Th1-biased response induced by an epitope vaccine composed of cholera toxin B unit and Helicobacter pylori Lpp20 epitopes. Helicobacter. 2016 [cited 15 September 2021];21(3):234-248 Available from: https://pubmed.ncbi.nlm.nih.gov/26332255/
  42. 42. Sutton P, Boag JM. Status of vaccine research and development for Helicobacter pylori. Vaccine. 2019 Nov;37(50):7295-7299. DOI: 10.1016/j.vaccine.2018.01.001
  43. 43. Malfertheiner P, Selgrad M, Wex T, Romi B, Borgogni E, Spensieri F, et al. Articles 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. The Lancet Gastroenterology & Hepatology. 2018 Oct;3(10):698-707 [cited 15 September 2021] Available from: www.thelancet.com/gastrohepPublishedonline
  44. 44. Malfertheiner P, Schultze V, Rosenkranz B, Kaufmann SH, Ulrichs T, Novicki D, Norelli F, Contorni M, Peppoloni S, Berti D, Tornese D, Ganju J, Palla E, Rappuoli R, Scharschmidt BF, Del Giudice G. Safety and immunogenicity of an intramuscular Helicobacter pylori vaccine in noninfected volunteers: A phase I study. Gastroenterology. 2008 [cited 15 september 2021];135(3):787-795 Available from: https://pubmed.ncbi.nlm.nih.gov/18619971/

Written By

Rike Syahniar, Dayu Swasti Kharisma and Rayhana

Submitted: 16 September 2021 Reviewed: 10 October 2021 Published: 26 November 2021