Next Article in Journal
Fabrication and Characterization of Celecoxib-Loaded Chitosan/Guar Gum-Based Hydrogel Beads
Next Article in Special Issue
Phenotypic and Genotypic Comparison of Antimicrobial-Resistant Variants of Escherichia coli and Salmonella Typhimurium Isolated from Evolution Assays with Antibiotics or Commercial Products Based on Essential Oils
Previous Article in Journal
A Novel Hydroxyapatite/Vitamin B12 Nanoformula for Treatment of Bone Damage: Preparation, Characterization, and Anti-Arthritic, Anti-Inflammatory, and Antioxidant Activities in Chemically Induced Arthritic Rats
Previous Article in Special Issue
CRISPR-Cas System: A Tool to Eliminate Drug-Resistant Gram-Negative Bacteria
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Potential of Alternative Therapies and Vaccine Candidates against Helicobacter pylori

1
Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia
2
Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
3
Department of Hematology and Oncology, Shenzhen Children’s Hospital, Shenzhen 518038, China
4
School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BA, UK
5
National Horizons Centre, Teesside University, Darlington DL1 1HG, UK
*
Authors to whom correspondence should be addressed.
Pharmaceuticals 2023, 16(4), 552; https://doi.org/10.3390/ph16040552
Submission received: 27 February 2023 / Revised: 31 March 2023 / Accepted: 31 March 2023 / Published: 6 April 2023

Abstract

:
Alternative therapies and vaccination are essential to combat the emergence of multidrug-resistant Helicobacter pylori and to prevent the development of gastroduodenal diseases. This review aimed to systematically review recent studies on alternative therapies, i.e., probiotics, nanoparticles, and natural products from plants, as well as recent progress in H. pylori vaccines at the preclinical stage. Articles published from January 2018 to August 2022 were systematically searched using PubMed, Scopus, Web of Science, and Medline. After the screening process, 45 articles were eligible for inclusion in this review. Probiotics (n = 9 studies) and natural products from plants (n = 28 studies) were observed to inhibit the growth of H. pylori, improve immune response, reduce inflammation, and reduce the pathogenic effects of H. pylori virulence factors. Natural products from plants also showed anti-biofilm activity against H. pylori. However, clinical trials of natural products from plants and probiotics are still lacking. A paucity of data assessing the nanoparticle activity of N-acylhomoserine lactonase-stabilized silver against H. pylori was observed. Nonetheless, one nanoparticle study showed anti-biofilm activity against H. pylori. Promising results of H. pylori vaccine candidates (n = 7) were observed at preclinical stage, including elicitation of a humoral and mucosal immune response. Furthermore, the application of new vaccine technology including multi-epitope and vector-based vaccines using bacteria was investigated at the preclinical stage. Taken together, probiotics, natural products from plants, and nanoparticles exhibited antibacterial activity against H. pylori. New vaccine technology shows promising results against H. pylori.

Graphical Abstract

1. Introduction

Helicobacter pylori infects more than 50% of the world’s population and causes gastroduodenal diseases including gastritis, peptic ulcer, gastric adenocarcinoma, and gastric lymphoma; it has been classified as a type I carcinogen that causes gastric cancer [1]. Gastric cancer is still one of the leading causes of cancer-related death worldwide although the prevalence and incidence of this cancer have been decreasing since the last decade [2]. Eradication of H. pylori is recommended to prevent gastric cancer, especially in developing countries where gastric cancer contributes to high economic morbidity and mortality [3]. Treatment of H. pylori includes administration of multiple antibiotics, namely, clarithromycin, amoxicillin, metronidazole, and tetracycline [4]. However, the emergence of H. pylori strains that are resistant to multiple antibiotics has complicated the treatment strategy to eradicate this bacterium [5]. In 2018, the World Health Organization listed H. pylori as one of the high-priority pathogens for research and discovery of novel drugs [6].
Multiple alternative therapies, including natural products, probiotics, and nanoparticles, can be assessed for antibacterial activity against H. pylori. A previous systematic review explored the efficacy of antimicrobial peptides natural sources as a promising alternative therapy for H. pylori [7]. Numerous natural products from plants have also been demonstrated to possess antibacterial activity against H. pylori, and they have been used as traditional medicine in some cultures such as East Asian and Southeast Asian cultures to treat multiple infections [8]. Probiotics are generally regarded as safe microbes that have been shown to give benefits to humans and are usually isolated from the fermentation process, including traditional foods [9]. In addition to probiotics and natural products from plants, applications of nanoparticles in drug delivery for antibiotics, implantable medical devices, and bone cement have also been explored for antibacterial effects against multiple bacteria [10]. Despite evidence demonstrating the efficacy of natural products from plants and probiotics against H. pylori, they are still not widely approved for therapy of H. pylori. Prevention of H. pylori infection through vaccination is also pertinent to prevent gastroduodenal diseases. To date, no vaccine for H. pylori has been approved. The objective of this review was to systematically review recent studies on alternative therapies (natural products from plants, probiotics, and nanoparticles) against H. pylori and progress of H. pylori vaccines.

2. Results

2.1. Literature Assessment

Figure 1 illustrates the screening process adopted for inclusion and exclusion of the articles in this systematic review. A total of 7796 articles were obtained from the following literature databases: PubMed (n = 852), Scopus (n = 743), Web of Science (n = 332), and EBSCO Medline (n = 5869). After we removed the duplicates (n = 1482) and reviews (n = 3319) using Microsoft Excel 2016 and Mendeley reference manager, 2995 articles were eligible for title and abstract screening. An additional 2745 articles were excluded because they were not relevant to the research question. Hence, 250 articles were eligible for full-text evaluation, of which 205 articles were excluded because the studies (n = 53) used natural products, probiotics, or nanoparticles in combination with commercial antibiotics, and the studies were published prior to 2018 (n = 152). Finally, 45 articles were eligible to be included in this systematic review. Overall, seven studies (15.5%) were published in 2018, along with 10 (22.2%) in 2019, 10 (22.2%) in 2020, 10 (22.2%) in 2021, and eight (17.8%) in 2022. Most of the studies published were preclinical studies (n = 44; 97.8%), while only one study (2.2%) was a clinical trial. Nine studies evaluated potential probiotics against H. pylori, 28 studies evaluated plant natural products against H. pylori, one study evaluated nanoparticles only, and seven studies evaluated vaccine candidates.

2.2. Summary of Studies on Alternative Therapies and Vaccines Conducted Prior to 2018

As the objective of our review was to systematically review recent studies published on alternative therapies and vaccines against H. pylori from 2018 to 2022, we excluded studies published prior to 2018. However, we briefly summarize breakthrough discoveries on this topic before 2018. According to our search, numerous studies on alternative therapies against H. pylori were published prior to 2018. Most of the studies examined the antibacterial activity of products from plants against H. pylori with promising results, including inhibition of growth in vitro, bacterial load reduction in animal models, and suppression of H. pylori virulence factors. Additionally, most studies were conducted at the preclinical level. Progress has been made in the discovery of vaccines against H. pylori, including a clinical trial conducted in China (NCT02302170), where they found the administration of oral recombinant H. pylori in children to be safe and effective in preventing H. pylori infection [11]. In probiotics research, one clinical trial using a combination of eight bacteria administered to 40 patients showed promising results, whereby H. pylori was eradicated in 13 patients [12]. In summary, we found potential clinical application of alternative therapies and vaccines against H. pylori before 2018.

2.3. Antibacterial Activity of Probiotics against H. pylori

Nine studies assessed the antibacterial activity of potential probiotics against H. pylori, of which eight were preclinical studies while one was a clinical trial (Table 1).
Most studies assessed the antibacterial activity of Lactobacillus spp. (8/9; 88.9%) against H. pylori, of which four studies determined the antibacterial properties of L. acidophilus, two studies determined the properties of Bifidobacterium spp., and one study each determined the properties of Streptococcus thermophilus and Parabacteroides goldsteinii MTS01. Of note, Lactobacillus spp. were demonstrated to show antibacterial activity against H. pylori in vitro and in vivo studies. Four studies showed that Lactobacillus spp., namely, L. casei, L. paracasei, L. acidophilus, L. rhamnosus, and L. fermentum, inhibited the growth of H. pylori in vitro. Notably, one study showed that L. rhamnosus and L. acidophilus inhibited the growth of multidrug-resistant H. pylori in vitro [14] (Table 1). S. thermophilus and B. lactis also inhibited the growth of H. pylori. Synthesis of data based on the antibacterial mechanism of probiotics against H. pylori revealed that most studies (n = 4) found the administration of probiotics in either animal models or humans altered the gut microbiota of the host infected with H. pylori. In one study, mice infected with H. pylori, but treated with probiotics, were shown to harbor enriched beneficial microbes that produced short fatty acid chains such as Bacteroides, Alloprevotella, and Oscellibacter and anti-inflammatory microbes (Faecalibaculum) [20]. However, in a clinical trial, probiotic monotherapy consisting of B. infantis, L. acidophilus, E. faecalis (>0.5 × 106 CFU/tablet), and B. cereus for 14 days was not beneficial to H. pylori-infected subjects as the therapy was not successful in reducing H. pylori burden in human’s stomach (ChiCTR1900024893) [15]. Three studies found that administration of probiotics reduced inflammation in the host, in which the expression of interleukin-8 (IL-8) and nuclear factor kappa B (NF-κB) decreased in H. pylori-infected cell lines or animal models. In addition, molecules essential in proinflammatory cellular signaling were reduced in the H. pylori-infected host model [20]. Administration of probiotics also reduced H. pylori cell adhesion, as observed in two studies [14,16]. An increase in cell apoptosis was observed in two studies [17,19]. Furthermore, probiotics treatment was also demonstrated to reduce the effect of vacuolating cytotoxin A (VacA) vacuolation in cells [18,21], as well as cytotoxin-associated gene A (CagA) translocation, phosphorylation, and the “hummingbird” cell-scattering effect [14,21]. Lin et al. [18] also found that treatment of probiotics reduced H. pylori colonization burden and stimulated the release of metabolites important in the immune response in mice.

2.4. Antibacterial Activity of Natural Products from Plants and Nanoparticles against H. pylori

Supplementary Table S1 [22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49] shows a list of plants used to examine the antibacterial activity of natural products from plants against H. pylori. In total, 28 studies examining the antibacterial activity of natural products against H. pylori were included in this review. Most studies (n = 15) assessed the antibacterial activity of plant extracts, while the remaining studies (n = 13) assessed isolated compounds from plants. The minimal inhibitory concentration (MIC) value was assessed in 20 studies, among which eight studies also assessed the minimal bactericidal concentration (MBC) value. Most studies that reported the MIC value (n = 16) adopted the broth microdilution assay to assess MIC value, whereas the remaining studies used the disc diffusion assay (n = 2) or both the disc diffusion assay and the agar dilution method (n = 2). Nineteen out of 20 studies (95%) providing the MIC value reported the susceptibility of H. pylori against the plant extracts or isolated compounds examined, while one study (1/20; 5%) reported no direct antibacterial activity. The lowest MIC value was 1.25–5 µg/mL in a study that examined the compound nimbolide isolated from Azadirachta indica [41]. In addition, this compound from a similar study also demonstrated an MBC against H. pylori ranging from 2.5 to 10 μg/mL. A high MIC value ≥500 µg/mL was reported in the studies that examined the antibacterial activity of the compound taxifolin from Mimusops balata fruit [25], the ethyl acetate fraction of Physalis alkekengi L. var. franchetii, and the dry extract of Libidibia ferrea. The antibacterial mechanisms of plant extracts or compounds were examined in 20 studies.
Overall, 10 categories of antibacterial mechanism were examined in the studies: (1) anti-biofilm, (2) anti-urease, (3) gastroprotection, (4) anti-inflammation, (5) effect on H. pylori virulence factors, (6) ATP leakage from H. pylori, (7) immune response, (8) H. pylori conversion from spiral to coccoid (inactive form), (9) cellular signaling, and (10) bacterial burden. Two studies found that the ethyl acetate fraction from Hibiscus rosa-sinensis red flower [29] and phylligenin, a compound isolated from flowering plant Forsythia [37], exhibited anti-biofilm activity against H. pylori. Meanwhile, four studies found that natural plant products possessed anti-urease activity, and six studies showed the gastroprotective properties of plant natural products. Furthermore, six studies demonstrated the anti-inflammatory activity of six different plant products. Five studies showed a reduction in H. pylori virulence factors, namely, cagA, vacA, ureA, flaA, and Omp18, while three studies showed a reduction in bacterial load and colonization. Two studies each demonstrated a reduction in cellular signaling important in H. pylori-induced carcinogenesis [30,48] and conversion of H. pylori from the active (spiral) to inactive coccoid form [29,42]. Li et al. [37] observed that phylligenin extracted from Forsythia induced ATP leakage in H. pylori and inhibited the mechanism of antibacterial resistance in the bacteria, as well as induced a good immune response.
It is suggested that to be considered as good antibacterial agent against H. pylori, the potential therapeutic candidate natural product must have antibacterial activity against not only the bacteria itself, but also its virulence factors that orchestrate gastric carcinogenesis. Taken together, the studies included in this systematic review (Table S1) demonstrated the application of natural products that can be used as future alternative therapies against H. pylori. Mechanisms targeting H. pylori examined in this study included the effects of the products from plants against important H. pylori virulence factors, namely, urease and CagA, which are both important for H. pylori to establish colonization in the human stomach due to its inhospitable environment. Furthermore, the effect of plant compounds in decreasing CagA activity is also important to prevent gastric carcinogenesis orchestrated by this oncoprotein [30,48]. Nevertheless, modeling of the natural products against specific target molecules of H. pylori is still lacking. The interaction of potential compounds with H. pylori using molecular docking and machine learning, coupled with in vitro and in vivo experiments, should be conducted in the future to elucidate the mechanism of antibacterial activity.
Nanoparticles have emerged alternative antibacterial agents against H. pylori because of their chemical properties that enable the attachment to and disturbance of the membrane, targeting bacterial DNA replication and transcription, as well as RNA translation [50]. In this review, only one study [51] examined the antibacterial activity of nanoparticles against H. pylori. The nanoparticles (namely, N-acylhomoserine lactonase-stabilized silver nanoparticles (AiiA-AgNPs)) inhibited the quorum sensing molecules of H. pylori, which in turn reduced the biofilm formation of the bacteria, production of urease, and cell surface hydrophobicity of H. pylori [51].

2.5. Progress on H. pylori Vaccine

Seven studies that evaluated vaccine candidates against H. pylori were included in this systematic review. A list of the vaccine candidates examined is available in Table 2.
All studies adopted mouse models to study the vaccine efficacy. The types of vaccine adopted in the studies included vector-based vaccines using vectors of Saccharomyces cerevisiae (n = 1), Listeria monocytogenes (n = 1), and Lactococcus lactis (n = 1). Multivalent epitope vaccines were employed in two studies, and outer membrane vesicles and peptide antigens were used in one study each. Six studies (85.7%) administered vaccine delivery to the animal models orally, while one study (14.3%) administered the vaccine through subcutaneous injection. H. pylori virulence factors targeted for vaccine design included urease, CagA, VacA, FlaA, neutrophil-activating protein A subunit, outer membrane vesicles, AlpB, SabA, and HpaA. Two studies that employed multivalent epitope vaccines targeted epitopes on B and T cells. A study conducted by Liu et al. [55] targeted 169 outer membrane proteins of H. pylori. In summary, all vaccine candidates showed promising results at a preclinical level. All studies observed higher IgA and IgG levels in mice immunized with vaccine candidates compared to controls. A reduction in the bacterial load and stomach inflammation were observed in most studies (6/7; 85.7%). Furthermore, three studies showed that immunization elicited an immune response in mouse models, including an increase in the secretion of interferon gamma (IFN-γ), IL-4, and IL-17. Contradictory T-cell polarization was observed, whereby one study [55] observed T helper 2 (Th2) polarization in immunized mice while Peng et al. [56] observed Th17 and Th1 polarization.

3. Discussion

In this systematic review, we systematically reviewed the antibacterial activity of probiotics, nanoparticles, and natural products from plants against H. pylori, as well as progress in H. pylori vaccine development. We restricted our analysis to articles published from 2018 to 2022 to obtain insights into recent publications describing alternative therapies against H. pylori. Studies that assessed the antibacterial activity of alternative therapies in combination with approved commercialized antibiotics were excluded from our review because the objective of our review was to assess the efficacy of alternative therapy administered as monotherapy. This is because antibiotics have been shown to disturb the gut microbiota and microbiome, which can have negative consequences for gastrointestinal health [59]. Additionally, antibiotic treatment regimens to eradicate H. pylori consist of multiple antibiotics, which may hinder patient compliance and contribute to an increase in the H. pylori antibiotic resistance rate. Hence, the discovery of alternative monotherapy is pertinent in the research and development of new drugs against H. pylori. Nevertheless, studies have shown that supplementation of probiotics together with antibiotics has helped to eradicate H. pylori infection in patients [60,61]. Despite an observed increase in publications from 2018 to 2022 regarding the alternative therapy of H. pylori in preclinical studies, a lack of publications on human studies was noted. Factors that contribute to a lack of clinical trials on potential therapies include a lack of financial resources and skilled medical specialists, as well as government regulations and administrative issues, specifically in middle- and low-income countries [62]. As the prevalence of H. pylori is high in middle- and low-income countries [63], collaboration between high-income countries and middle- and low-income countries in clinical trials should be encouraged to mitigate this issue. The studies included in this systematic review showed that some probiotics exhibited antibacterial activity or competitive exclusion against H. pylori. Furthermore, antibacterial mechanisms of probiotics against H. pylori, including effects on virulence factors, the gut microbiota, and the immune response, were also explored. However, there was a lack of studies that identified and assessed the antibacterial activity of compounds secreted by probiotics against H. pylori. This gap should be examined in future studies. Of note, the clinical trial included in this review that investigated the effect of probiotic monotherapy on human subjects failed to show a beneficial effect of probiotics in human subjects. In contrast, administration of probiotics together with antibiotics was demonstrated to improve eradication with minimal adverse effects [64]. Thus, the mechanism (i.e., drug synergism and antagonism of probiotic monotherapy and probiotic supplementation with antibiotics) of combating H. pylori infection in human should be further investigated in the future.
A promising application of natural products from plants was also observed in this systematic review. Every study adopted different plant species to determine antibacterial activity, suggesting a broad spectrum of plant types that can be used as therapy against the bacterium. The antibacterial activity of plant species against H. pylori has mostly been determined from plant extracts without an identification of specific compounds with activity against H. pylori. Given the complexity of plant chemical constituents, isolation of desired chemical compound from plants is important to determine the safety and the antibacterial efficacy of the compound intended for future human study. Most studies reported MIC values of the products examined. Nevertheless, a lack of reports including MBC values was noted. MIC values only provide information on the growth inhibition of H. pylori, while MBC value provide information on bacteria killing. Studies to determine antibacterial activity in the future should also report the MBC value of H. pylori. The Clinical and Laboratory Standard Institute (CLSI) recommends the agar dilution method to determine the MIC value of H. pylori [65]. However, we observed that most studies employed the broth microdilution assay to determine the MIC value. This discrepancy is because the agar dilution assay is difficult to perform and laborious, in addition to the fact that H. pylori grows slowly. Various antibacterial mechanisms of plant products against H. pylori were examined, including effects on virulence factors, bacterial burden in the host, and anti-biofilm activity. Although the emergence of multidrug-resistant H. pylori has been observed in Southeast Asia [7], there is a paucity of recent studies examining the antibacterial activity of plant natural products in this region. Given the richness of tropical biodiversity in this region, the anti-H. pylori activities of products from plants should be explored in the future using the diverse plant species in the region. Nanoparticles have been demonstrated to display antimicrobial activity against multiple types of bacteria [66]. Nevertheless, only one recent study assessed the antibacterial activity of nanoparticles against H. pylori with promising results. Barriers to the application of nanoparticles as antibacterial agents include the design of nanoparticles for efficient delivery to the host and the toxicity of the chemical to the host [67].
Vaccination remains a strategy to prevent infection. To date, no approved H. pylori vaccine is available. Nevertheless, phase III clinical trials of an H. pylori oral vaccine in 4464 participants were conducted with effective, safe, and immunogenic results [11]. All studies conducted at the preclinical stage in this systematic review demonstrated the promising results of H. pylori vaccine candidates, in which the host’s immune response was elicited in all studies. Interestingly, most studies explored the application of new vaccine technologies including vector-based vaccines using bacteria and multi-epitope vaccines targeting specific T and B cells. The unique design of multi-epitope vaccines harnesses a better immune response than single-epitope vaccines, particularly for H. pylori strains with different antigen variability [68]. Meanwhile, vector-based vaccines provide a robust immune response since live bacteria are easily recognized by the immune system, and since the delivery of live bacteria is more efficient compared to traditional vaccines [69]. Contradictory results regarding T-cell polarization have been found, whereby Peng et al. [56] found Th1 to be polarized in H. pylori vaccination, while Liu et al. [55] found Th2 to be polarized. Th2 cells are important to neutralize extracellular bacteria such as H. pylori, while Th1 cells are important to neutralize intracellular bacteria [70]. However, both studies showed promising results of H. pylori vaccine immunization. Further studies should be conducted to elucidate the importance of Th1 and Th2 cells in H. pylori immunization.
While the data obtained from the studies included in this systematic review that examined the alternative therapies against H. pylori are encouraging, there were several limitations in this review. Firstly, we noticed heterogeneity of the studies included in this systematic review. The methods adopted to examine antibacterial activity were not universal, whereby some studies used broth microdilution while other studies used an agar diffusion assay. However, the difficulty in culturing H. pylori in the laboratory with different types of plants contributed to the choice of selecting appropriate antibacterial assays that differ from one laboratory to another laboratory. Secondly, we only included studies that were published in English, which may have missed studies that were published in other languages. Lastly, most studies included in this review involved preclinical trials, thus hampering our understanding of whether the findings from preclinical studies can be translated to clinical trials. The lack of studies conducted on humans may have stemmed from the expensive cost to conduct clinical trials, especially in developing countries with limited funding resources and experts in respective fields. In addition, a lack of financial resources in developing countries is also one of the factors leading to a lack of product patents in preclinical studies that can be used for clinical trials before commercialization. Lack of expertise in managing clinical trials is also a challenge that developing countries must face before the implementation of clinical trials. Hence, this factor has contributed to a lack of clinical trials conducted in developing countries where H. pylori infection is high.

4. Materials and Methods

4.1. Literature Search

Four literature databases, namely, PubMed, Scopus, Web of Science, and EBSCO Medline, were used for literature search. We systematically evaluated all articles obtained from the literature search using Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The following keywords were used to search the articles: ((“natural product” OR “plant” OR “nanoparticle” OR “probiotic” OR “vaccine”) AND (“treatment”) AND (“Helicobacter pylori” OR “H. pylori”)). We defined natural products as compounds or extracts obtained from plant sources because our systematic review focused on natural products from plants. For probiotics, we defined the term as live microbes generally regarded as safe and administered to hosts that may have benefits for the hosts. Commercialized antibiotics are defined as antibiotics currently available to treat H. pylori, which include amoxicillin, tetracycline, clarithromycin, metronidazole, levofloxacin and rifampicin. Nanoparticles are ultrasmall particles in the range of 1–100 nm in diameter, while vaccines are defined as substances that can elicit an immune system response against H. pylori.

4.2. Inclusion and Exclusion Criteria

Inclusion criteria of this study included (1) studies that evaluated natural products from plants or nanoparticles against H. pylori, (2) studies that evaluated the antibacterial properties of potential probiotics against H. pylori, (3) studies that evaluated vaccine candidates of H. pylori in preclinical trials, (4) studies that were published in the last 5 years (from January 2018 to August 2022), (5) studies conducted in vitro, in vivo, or in humans, and (6) studies published in English with the full text available. Exclusion criteria included (1) studies that evaluated natural products, potential probiotics, or nanoparticles in combination with commercialized antibiotics against H. pylori, (2) studies that evaluated potential probiotics, natural products, vaccine, or nanoparticles against bacteria other than H. pylori, (3) studies not published in English or with full text not available, (4) studies that examined vaccine candidates in clinical trials, (5) book chapters, conference abstracts, and literature, systematic, or meta-analysis reviews, and (6) studies published before 2018. Two independent researchers evaluated the articles to be included in this review; if there was disagreement for inclusion or exclusion of the articles, a third researcher was consulted to reach consensus. The literature search was conducted from May to August 2022.

4.3. Data Extraction and Synthesis

Data such as authors, year, type of study (preclinical or clinical trials), type of natural products used in the study, name of probiotic or name of vaccine candidate and their evaluation in vitro and in vivo, and results of antibacterial activity against H. pylori were extracted and organized using Microsoft Excel 2016. Data were synthesized according to antibacterial activity and mechanism of the products against H. pylori.

5. Conclusions

In conclusion, probiotics and natural products from plants show promising results to be harnessed as alternative therapies against H. pylori to combat the emergence of multidrug-resistant strains. However, there is still a paucity of clinical trials on probiotics and natural products from plants. This stems from the fact that H. pylori infections are mostly diagnosed in developing countries where clinical trials are expensive and require a team of clinical experts. Consequently, the findings from preclinical studies cannot be properly translated to human studies. With the advent of artificial intelligence and machine learning technology, the research and development of drugs and vaccines are no longer limited to wet laboratory experiments; they can also include silico studies. Of note, studies that employed both wet and dry laboratory experiments for drug discovery were still lacking in our systematic review. In silico studies prior to commencement of wet laboratory experiments can provide valuable insights into potential drug candidates that can be screened in vitro and in vivo. Probiotics from fermented foods and natural products extracted from plants, especially from areas with rich biodiversity, can be harnessed for the research and discovery of novel antimicrobial agents against antibiotic-resistant H. pylori. H. pylori vaccines using new vaccine technologies show promising results in preclinical trials and should be explored further.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ph16040552/s1: Table S1. List of natural products from plants described in the studies included in this systematic review.

Author Contributions

Conceptualization, A.S. and A.H.; methodology, A.S., B.S.L., and S.P.; validation, A.H., B.S.L., and S.P.; formal analysis, A.S.; data curation, A.S.; writing—original draft preparation, A.S.; writing—review and editing, B.S.L. and A.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Ministry of Higher Education of Malaysia, grant no. FRGS/1/2019/SKK11/UKM/02/3.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data from this review are available from the corresponding authors upon reasonable request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Alexander, S.M.; Retnakumar, R.J.; Chouhan, D.; Devi, T.N.B.; Dharmaseelan, S.; Devadas, K.; Thapa, N.; Tamang, J.P.; Lamtha, S.C.; Chattopadhyay, S. Helicobacter pylori in human stomach: The inconsistencies in clinical outcomes and the probable causes. Front. Microbiol. 2021, 12, 713955. [Google Scholar] [CrossRef]
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  3. Ford, A.C.; Yuan, Y.; Moayyedi, P. Helicobacter pylori eradication therapy to prevent gastric cancer: Systematic review and meta-analysis. Gut 2020, 69, 2113–2121. [Google Scholar] [CrossRef]
  4. Goderska, K.; Agudo Pena, S.; Alarcon, T. Helicobacter pylori treatment: Antibiotics or probiotics. Appl. Microbiol. Biotechnol. 2018, 102, 1–7. [Google Scholar] [CrossRef]
  5. Savoldi, A.; Carrara, E.; Graham, D.Y.; 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, 1372–1382.e17. [Google Scholar] [CrossRef] [Green Version]
  6. Tacconelli, E.; Carrara, E.; Savoldi, A.; Harbarth, S.; Mendelson, M.; Monnet, D.L.; Pulcini, C.; Kahlmeter, G.; Kluytmans, J.; Carmeli, Y.; et al. Discovery, research, and development of new antibiotics: The WHO priority list of antibiotic-resistant bacteria and tuberculosis. Lancet Infect. Dis. 2018, 18, 318–327. [Google Scholar] [CrossRef]
  7. Sukri, A.; Lopes, B.S.; Hanafiah, A. The emergence of multidrug-resistant Helicobacter pylori in Southeast Asia: A systematic review on the trends and intervention strategies using antimicrobial peptides. Antibiotics 2021, 10, 1061. [Google Scholar] [CrossRef]
  8. Chassagne, F.; Samarakoon, T.; Porras, G.; Lyles, J.T.; Dettweiler, M.; Marquez, L.; Salam, A.M.; Shabih, S.; Farrokhi, D.R.; Quave, C.L. A systematic review of plants with antibacterial activities: A taxonomic and phylogenetic perspective. Front. Pharmacol. 2021, 11, 586548. [Google Scholar] [CrossRef]
  9. Bagchi, T. Traditional food & modern lifestyle: Impact of probiotics. Indian J. Med. Res. 2014, 140, 333–335. [Google Scholar]
  10. Wang, L.; Hu, C.; Shao, L. The antimicrobial activity of nanoparticles: Present situation and prospects for the future. Int. J. Nanomed. 2017, 12, 1227–1249. [Google Scholar] [CrossRef] [Green Version]
  11. Zeng, M.; Mao, X.H.; Li, J.X.; Tong, W.D.; Wang, B.; Zhang, Y.J.; Guo, G.; Zhao, Z.J.; Li, L.; Wu, D.L.; 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, 1457–1464. [Google Scholar] [CrossRef]
  12. Rosania, R.; Minenna, M.F.; Giorgio, F.; Facciorusso, A.; De Francesco, V.; Hassan, C.; Panella, C.; Ierardi, E. Probiotic multistrain treatment may eradicate Helicobacter pylori from the stomach of dyspeptics: A placebo-controlled pilot study. Inflamm. Allergy Drug Targets 2012, 11, 244–249. [Google Scholar] [CrossRef]
  13. Saracino, I.M.; Pavoni, M.; Saccomanno, L.; Fiorini, G.; Pesci, V.; Foschi, C.; Piccirilli, G.; Bernardini, G.; Holton, J.; Figura, N.; et al. Antimicrobial efficacy of five probiotic strains against Helicobacter pylori. Antibiotics 2020, 9, 244. [Google Scholar] [CrossRef]
  14. Chen, Y.H.; Tsai, W.H.; Wu, H.Y.; Chen, C.Y.; Yeh, W.L.; Chen, Y.H.; Hsu, H.Y.; Chen, W.W.; Chen, Y.W.; Chang, W.W.; et al. Probiotic Lactobacillus spp. act against Helicobacter pylori-induced inflammation. J. Clin. Med. 2019, 8, 90. [Google Scholar] [CrossRef] [Green Version]
  15. Yuan, Z.; Xiao, S.; Li, S.; Suo, B.; Wang, Y.; Meng, L.; Liu, Z.; Yin, Z.; Xue, Y.; Zhou, L. The impact of Helicobacter pylori infection, eradication therapy, and probiotics intervention on gastric microbiota in young adults. Helicobacter 2021, 26, e12848. [Google Scholar] [CrossRef]
  16. Taghizadeh, S.; Falsafi, T.; Kermanshahi, R.K.; Ramezani, R. Antagonistic and Immunomodulant effects of two probiotic strains of Lactobacillus on clinical strains of Helicobacter pylori. Galen. Med. J. 2020, 9, e1794. [Google Scholar] [CrossRef]
  17. Yarmohammadi, M.; Yadegar, A.; Ebrahimi, M.T.; Zali, M.R. Effects of a potential probiotic strain Lactobacillus gasseri ATCC 33323 on Helicobacter pylori-induced inflammatory response and gene expression in coinfected gastric epithelial cells. Probiotics Antimicrob. Proteins 2021, 13, 751–764. [Google Scholar] [CrossRef]
  18. Lin, C.C.; Huang, W.C.; Su, C.H.; Lin, W.D.; Wu, W.T.; Yu, B.; Hsu, Y.M. Effects of multi-strain probiotics on immune responses and metabolic balance in Helicobacter pylori-infected mice. Nutrients 2020, 12, 2476. [Google Scholar] [CrossRef]
  19. Maleki-Kakelar, H.; Dehghani, J.; Barzegari, A.; Barar, J.; Shirmohamadi, M.; Sadeghi, J.; Omidi, Y. Lactobacillus plantarum induces apoptosis in gastric cancer cells via modulation of signaling pathways in Helicobacter pylori. BioImpacts 2020, 10, 65–72. [Google Scholar] [CrossRef] [Green Version]
  20. He, C.; Peng, C.; Xu, X.; Li, N.; Ouyang, Y.; Zhu, Y.; Lu, N. Probiotics mitigate Helicobacter pylori-induced gastric inflammation and premalignant lesions in INS-GAS mice with the modulation of gastrointestinal microbiota. Helicobacter 2022, 27, e12898. [Google Scholar] [CrossRef]
  21. Lai, C.H.; Lin, T.L.; Huang, M.Z.; Li, S.W.; Wu, H.Y.; Chiu, Y.F.; Yang, C.Y.; Chiu, C.H.; Lai, H.C. Gut commensal Parabacteroides goldsteinii MTS01 alters gut microbiota composition and reduces cholesterol to mitigate Helicobacter pylori-Induced Pathogenesis. Front. Immunol. 2022, 13, 916848. [Google Scholar] [CrossRef]
  22. Eftekhari, M.; Shams Ardekani, M.R.; Amin, M.; Mansourian, M.; Saeedi, M.; Akbarzadeh, T.; Khanavi, M. Anti-Helicobacter pylori compounds from Oliveria decumbens Vent. through urease inhibitory in-vitro and in-silico studies. Iran. J. Pharm. Res. 2021, 20, 476–489. [Google Scholar]
  23. Palacios-Espinosa, J.F.; Núñez-Aragón, P.N.; Gomez-Chang, E.; Linares, E.; Bye, R.; Romero, I. Anti-Helicobacter pylori activity of Artemisia ludoviciana subsp. mexicana and two of its bioactive components, estafiatin and eupatilin. Molecules 2021, 26, 3654. [Google Scholar] [CrossRef]
  24. Lee, H.A.; Hong, S.; Yoo, J.H.; Chung, Y.; Kim, O. Anti-Helicobacter pylori activity and inhibition of gastritis by Allium hookeri extract. Lab. Anim. Res. 2018, 34, 75–79. [Google Scholar] [CrossRef] [Green Version]
  25. Stenger Moura, F.C.; Cechinel-Filho, V.; Greco, F.A.; Venzon, L.; Meurer, M.C.; França, T.C.D.S.; Longo, B.; Somensi, L.B.; Mariano, L.N.B.; Cruz, A.B.; et al. Taxifolin and gastro-adhesive microparticles containing taxifolin promotes gastric healing in vivo, inhibits Helicobacter pylori in vitro and proton pump reversibly in silico. Chem. Biol. Interact. 2021, 339, 109445. [Google Scholar] [CrossRef]
  26. Lu, Q.; Li, C.; Wu, G. Insight into the inhibitory effects of Zanthoxylum nitidum against Helicobacter pylori urease and jack bean urease: Kinetics and mechanism. J. Ethnopharmacol. 2020, 249, 112419. [Google Scholar] [CrossRef]
  27. Ngnameko, C.R.; Marchetti, L.; Zambelli, B.; Quotadamo, A.; Roncarati, D.; Bertelli, D.; Njayou, F.N.; Smith, S.I.; Moundipa, P.F.; Costi, M.P.; et al. New insights into bioactive compounds from the medicinal plant Spathodea campanulata P. Beauv. and their activity against Helicobacter pylori. Antibiotics 2020, 9, 258. [Google Scholar]
  28. Park, H.S.; Wijerathne, C.U.B.; Jeong, H.Y.; Seo, C.S.; Ha, H.; Kwun, H.J. Gastroprotective effects of Hwanglyeonhaedok-tang against Helicobacter pylori-induced gastric cell injury. J. Ethnopharmacol. 2018, 216, 239–250. [Google Scholar] [CrossRef]
  29. Ngan, L.T.M.; Tan, M.T.; Hoang, N.V.M.; Thanh, D.T.; Linh, N.T.T.; Hoa, T.T.H.; Nuong, N.T.M.; Hieu, T.T. Antibacterial activity of Hibiscus rosa-sinensis L. red flower against antibiotic-resistant strains of Helicobacter pylori and identification of the flower constituents. Brazil. J. Med. Biol. Res. 2021, 54, e10889. [Google Scholar] [CrossRef]
  30. Park, B.; Lim, J.W.; Kim, H. Lycopene treatment inhibits activation of Jak1/Stat3 and Wnt/β-catenin signaling and attenuates hyperproliferation in gastric epithelial cells. Nutr. Res. 2019, 70, 70–81. [Google Scholar] [CrossRef]
  31. Salinas Ibáñez, Á.G.; Vallés, D.; Adaro, M.; Barberis, S.; Vega, A.E. Antimicrobial effect of a proteolytic enzyme from the fruits of Solanum granuloso-leprosum (Dunal) against Helicobacter pylori. Front. Nutr. 2021, 8, 699955. [Google Scholar] [CrossRef] [PubMed]
  32. Jin, H.F.; Dai, J.F.; Meng, L.N.; Lu, B. Curcuma wenyujin Y. H. Chen et C. Ling n-butyl alcohol extract inhibits AGS cell Helicobacter pylori CagA+VacA+ promoted invasiveness by down-regulating caudal type homeobox transcription factor and claudin-2 expression. Chin. J. Integr. Med. 2020, 26, 122–129. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, Y.; Wang, S.L.; Zhang, J.Y.; Song, X.N.; Zhang, Z.Y.; Li, J.F.; Li, S. Anti-ulcer and anti-Helicobacter pylori potentials of the ethyl acetate fraction of Physalis alkekengi L. var. franchetii (Solanaceae) in rodent. J. Ethnopharmacol. 2018, 211, 197–206. [Google Scholar] [CrossRef]
  34. Fahmy, N.M.; Al-Sayed, E.; Michel, H.E.; El-Shazly, M.; Singab, A.N.B. Gastroprotective effects of Erythrina speciosa (Fabaceae) leaves cultivated in Egypt against ethanol-induced gastric ulcer in rats. J. Ethnopharmacol. 2020, 248, 112297. [Google Scholar] [CrossRef]
  35. Lee, Y.I.; Kim, J.S.; Cho, J.S.; Kim, H.K.; Hussain, A. Standardized combined plant extract, RUG-com, reduces bacterial levels and suppresses acute and chronic inflammation in Balb/c mice infected with CagA+ Helicobacter pylori. Prev. Nutr. Food Sci. 2019, 24, 426–433. [Google Scholar] [CrossRef] [PubMed]
  36. Jung, D.H.; Park, M.H.; Kim, C.J.; Lee, J.Y.; Keum, C.Y.; Kim, I.S.; Yun, C.H.; Kim, S.K.; Kim, W.H.; Lee, Y.C. Effect of β-caryophyllene from cloves extract on Helicobacter pylori eradication in mouse model. Nutrients 2020, 12, 1000. [Google Scholar] [CrossRef] [Green Version]
  37. Li, R.J.; Qin, C.; Huang, G.R.; Liao, L.J.; Mo, X.Q.; Huang, Y.Q. Phillygenin inhibits Helicobacter pylori by preventing biofilm formation and inducing ATP leakage. Front. Microbiol. 2022, 13, 863624. [Google Scholar] [CrossRef] [PubMed]
  38. Eftekhari, M.; Shams Ardekani, M.R.; Amin, M.; Attar, F.; Akbarzadeh, T.; Safavi, M.; Karimpour-Razkenari, E.; Amini, M.; Isman, M.; Khanavi, M. Oliveria decumbens, a bioactive essential oil: Chemical composition and biological activities. Iran. J. Pharm. Res. 2019, 18, 412–421. [Google Scholar]
  39. Brito, S.A.; de Almeida, C.L.F.; de Santana, T.I.; da Silva Oliveira, A.R.; do Nascimento Figueiredo, J.C.B.; Souza, I.T.; de Almeida, L.L.; da Silva, M.V.; Borges, A.S.; de Medeiros, J.W.; et al. Antiulcer activity and potential mechanism of action of the leaves of Spondias mombin L. Oxid. Med. Cell Longev. 2018, 2018, 1731459. [Google Scholar] [CrossRef] [Green Version]
  40. Prazeres, L.D.K.T.; Aragão, T.P.; Brito, S.A.; Almeida, C.L.F.; Silva, A.D.; de Paula, M.M.F.; Farias, J.S.; Vieira, L.D.; Damasceno, B.P.G.L.; Rolim, L.A.; et al. Antioxidant and antiulcerogenic activity of the dry extract of pods of Libidibia ferrea Mart. ex Tul. (Fabaceae). Oxid. Med. Cell Longev. 2019, 2019, 1983137. [Google Scholar] [CrossRef] [Green Version]
  41. Wylie, M.R.; Windham, I.H.; Blum, F.C.; Wu, H.; Merrell, D.S. In vitro antibacterial activity of nimbolide against Helicobacter pylori. J. Ethnopharmacol. 2022, 285, 114828. [Google Scholar] [CrossRef] [PubMed]
  42. Peng, C.; Sang, S.; Shen, X.; Zhang, W.; Yan, J.; Chen, P.; Jiang, C.; Yuan, Y.; Zhu, W.; Yao, M. In vitro anti-Helicobacter pylori activity of Syzygium aromaticum and the preliminary mechanism of action. J. Ethnopharmacol. 2022, 288, 114995. [Google Scholar] [CrossRef]
  43. Sabry, M.M.; El-Fishawy, A.M.; El-Rashedy, A.A.; El Gedaily, R.A. Phytochemical investigation of Cordia africana Lam. stem bark: Molecular simulation approach. Molecules 2022, 27, 4039. [Google Scholar] [CrossRef]
  44. Almeida, G.V.B.; Arunachalam, K.; Balogun, S.O.; Pavan, E.; Ascêncio, S.D.; Soares, I.M.; Zanatta, A.C.; Vilegas, W.; Macho, A.; Oliveira Martins, D.T. Chemical characterization and evaluation of gastric antiulcer properties of the hydroethanolic extract of the stem bark of Virola elongata (Benth.) Warb. J. Ethnopharmacol. 2019, 231, 113–124. [Google Scholar] [CrossRef]
  45. So, H.M.; Yu, J.S.; Khan, Z.; Subedi, L.; Ko, Y.J.; Lee, I.K.; Park, W.S.; Chung, S.J.; Ahn, M.J.; Kim, S.Y.; et al. Chemical constituents of the root bark of Ulmus davidiana var. japonica and their potential biological activities. Bioorg. Chem. 2019, 91, 103145. [Google Scholar]
  46. Sreeja, P.S.; Arunachalam, K.; Saikumar, S.; Kasipandi, M.; Dhivya, S.; Murugan, R.; Parimelazhagan, T. Gastroprotective effect and mode of action of methanol extract of Sphenodesme involucrata var. paniculata (C.B. Clarke) Munir (Lamiaceae) leaves on experimental gastric ulcer models. Biomed. Pharmacother. 2018, 97, 1109–1118. [Google Scholar]
  47. de Cássia Dos Santos, R.; Bonamin, F.; Périco, L.L.; Rodrigues, V.P.; Zanatta, A.C.; Rodrigues, C.M.; Sannomiya, M.; Dos Santos Ramos, M.A.; Bonifácio, B.V.; Bauab, T.M.; et al. Juss partitions promote gastroprotection against peptic ulcers and improve healing through antioxidant and anti-inflammatory activities. Biomed. Pharmacother. 2019, 111, 1112–1123. [Google Scholar]
  48. Abdel-Baki, P.M.; El-Sherei, M.M.; Khaleel, A.E.; Abdel-Aziz, M.M.; Okba, M.M. Irigenin, a novel lead from Iris confusa for management of Helicobacter pylori infection with selective COX-2 and HpIMPDH inhibitory potential. Sci. Rep. 2022, 12, 11457. [Google Scholar] [CrossRef] [PubMed]
  49. Kim, H.S.; Lim, J.W.; Kim, H. Korean red ginseng extract inhibits IL-8 expression via Nrf2 activation in Helicobacter pylori-infected gastric epithelial cells. Nutrients 2022, 14, 1044. [Google Scholar] [CrossRef] [PubMed]
  50. Gupta, A.; Mumtaz, S.; Li, C.H.; Hussain, I.; Rotello, V.M. Combatting antibiotic-resistant bacteria using nanomaterials. Chem. Soc. Rev. 2019, 48, 415–427. [Google Scholar] [CrossRef] [PubMed]
  51. Gopalakrishnan, V.; Masanam, E.; Ramkumar, V.S.; Baskaraligam, V.; Selvaraj, G. Influence of N-acylhomoserine lactonase silver nanoparticles on the quorum sensing system of Helicobacter pylori: A potential strategy to combat biofilm formation. J. Basic Microbiol. 2020, 60, 207–215. [Google Scholar] [CrossRef]
  52. Cen, Q.; Gao, T.; Ren, Y.; Lu, X.; Lei, H. Immune evaluation of a Saccharomyces cerevisiae-based oral vaccine against Helicobacter pylori in mice. Helicobacter 2021, 26, e12772. [Google Scholar] [CrossRef] [PubMed]
  53. Wang, S.; Ma, J.; Ji, Q.; Liu, Q. Evaluation of an attenuated Listeria monocytogenes as a vaccine vector to control Helicobacter pylori infection. Immunol. Lett. 2021, 238, 68–74. [Google Scholar] [CrossRef] [PubMed]
  54. Xie, W.; Zhao, W.; Zou, Z.; Kong, L.; Yang, L. Oral multivalent epitope vaccine, based on UreB, HpaA, CAT, and LTB, for prevention and treatment of Helicobacter pylori infection in C57BL/6 mice. Helicobacter 2021, 26, e12807. [Google Scholar] [CrossRef]
  55. Liu, Q.; Li, X.; Zhang, Y.; Song, Z.; Li, R.; Ruan, H.; Huang, X. Orally-administered outer-membrane vesicles from Helicobacter pylori reduce H. pylori infection via Th2-biased immune responses in mice. Pathog. Dis. 2019, 77, ftz050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Peng, X.; Zhang, R.; Duan, G.; Wang, C.; Sun, N.; Zhang, L.; Chen, S.; Fan, Q.; Xi, Y. Production and delivery of Helicobacter pylori NapA in Lactococcus lactis and its protective efficacy and immune modulatory activity. Sci. Rep. 2018, 8, 6435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Espinosa-Ramos, D.; Caballero-Hernández, D.; Gomez-Flores, R.; Trejo-Chávez, A.; Pérez-Limón, L.J.; de la Garza-Ramos, M.A.; Tamez-Guerra, R.; Tamez-Guerra, P.; Rodriguez-Padilla, C. Immunization with a synthetic Helicobacter pylori peptide induces secretory IgA antibodies and protects mice against infection. Can. J. Infect. Dis. Med. Microbiol. 2019, 2019, 8595487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Pan, X.; Ke, H.; Niu, X.; Li, S.; Lv, J.; Pan, L. Protection against Helicobacter pylori infection in BALB/c mouse model by oral administration of multivalent epitope-based vaccine of cholera toxin b subunit-HUUC. Front. Immunol. 2018, 9, 1003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Ramirez, J.; Guarner, F.; Bustos Fernandez, L.; Maruy, A.; Sdepanian, V.L.; Cohen, H. Antibiotics as major disruptors of gut microbiota. Front. Cell Infect. Microbiol. 2020, 10, 572912. [Google Scholar] [CrossRef]
  60. Srinarong, C.; Siramolpiwat, S.; Wongcha-um, A.; Mahachai, V.; Vilaichone, R.K. Improved eradication rate of standard triple therapy by adding bismuth and probiotic supplement for Helicobacter pylori treatment in Thailand. Asian Pac. J. Cancer Prev. 2014, 15, 9909–9913. [Google Scholar] [CrossRef] [Green Version]
  61. Tongtawee, T.; Dechsukhum, C.; Leeanansaksiri, W.; Kaewpitoon, S.; Kaewpitoon, N.; Loyd, R.A.; Matrakool, L.; Panpimanmas, S. Effect of pretreatment with Lactobacillus delbrueckii and Streptococcus thermophillus on tailored triple therapy for Helicobacter pylori eradication: A prospective randomized controlled clinical trial. Asian Pac. J. Cancer Prev. 2015, 16, 4885–4890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Alemayehu, C.; Mitchell, G.; Nikles, J. Barriers for conducting clinical trials in developing countries—A systematic review. Int. J. Equity Health 2018, 17, 37. [Google Scholar] [CrossRef]
  63. Hooi, J.K.Y.; Lai, W.Y.; Ng, W.K.; Suen, M.M.Y.; Underwood, F.E.; Tanyingoh, D.; Malfertheiner, P.; Graham, D.Y.; Wong, V.W.S.; Wu, J.C.Y.; et al. Global prevalence of Helicobacter pylori infection: Systematic review and meta-analysis. Gastroenterology 2017, 153, 420–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Lü, M.; Yu, S.; Deng, J.; Yan, Q.; Yang, C.; Xia, G.; Zhou, X. Efficacy of probiotic supplementation therapy for Helicobacter pylori eradication: A meta-analysis of randomized controlled trials. PLoS ONE 2016, 11, e0163743. [Google Scholar] [CrossRef] [Green Version]
  65. CLSI. Methods for Antimicrobial Dilution and Disk Susceptibility Testing of Infrequently Isolated or Fastidious Bacteria, 3rd ed.; CLSI guideline M45; Clinical and Laboratory Standards Institute: Wayne, PA, USA, 2016. [Google Scholar]
  66. Sharmin, S.; Rahaman, M.M.; Sarkar, C.; Atolani, O.; Islam, M.T.; Adeyemi, O.S. Nanoparticles as antimicrobial and antiviral agents: A literature-based perspective study. Heliyon 2021, 7, e06456. [Google Scholar] [CrossRef]
  67. Staroń, A.; Długosz, O. Antimicrobial properties of nanoparticles in the context of advantages and potential risks of their use. J. Environ. Sci. Health A Tox. Hazard. Subst. Environ. Eng. 2021, 56, 680–693. [Google Scholar] [CrossRef] [PubMed]
  68. Zhang, L. Multi-epitope vaccines: A promising strategy against tumors and viral infections. Cell Mol. Immunol. 2018, 15, 182–184. [Google Scholar] [CrossRef] [Green Version]
  69. Yurina, V. Live bacterial vectors-a promising DNA vaccine delivery system. Med. Sci. 2018, 6, 27. [Google Scholar] [CrossRef] [Green Version]
  70. Larussa, T.; Leone, I.; Suraci, E.; Imeneo, M.; Luzza, F. Helicobacter pylori and T helper cells: Mechanisms of immune escape and tolerance. J. Immunol. Res. 2015, 2015, 981328. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Flow diagram outlining article screening for inclusion in this systematic review using PRISMA guidelines. After screening evaluation, 45 articles were eligible for inclusion in this systematic review.
Figure 1. Flow diagram outlining article screening for inclusion in this systematic review using PRISMA guidelines. After screening evaluation, 45 articles were eligible for inclusion in this systematic review.
Pharmaceuticals 16 00552 g001
Table 1. List of potential probiotics and their antibacterial activity against H. pylori included in this systematic review. UBT: urea breath test; LAB: lactic acid bacteria.
Table 1. List of potential probiotics and their antibacterial activity against H. pylori included in this systematic review. UBT: urea breath test; LAB: lactic acid bacteria.
AuthorsType of StudyName of Bacteria (Probiotics)Results
Saracino et al. (2020) [13]PreclinicalL. casei, L. paracasei, L. acidophilus, B. lactis, and S. thermophilusGrowth inhibition of H. pylori.
Chen et al. (2019) [14]PreclinicalL. rhamnosus and L. acidophilusInhibited growth, adhesion, and invasion of H. pylori; reduced H. pylori-induced inflammation (decreased NF-κB activity and IL-8 secretion); downregulated phosphorylation and translocation of CagA; reshaped gut microbiota.
Yuan et al. (2021) [15]ClinicalProbiotics therapy (Bifidobacterium tetravaccine tablets) included B. infantis > 0.5 × 106 CFU/tablet, L. acidophilus > 0.5 × 106 CFU/tablet, E. faecalis > 0.5 × 106 CFU/tablet, B. cereus > 0.5 × 105 CFU/tablet)Upregulated pathogenic bacteria in gut microbiota after administration of probiotics.
Taghizadeh et al. (2020) [16]PreclinicalL. acidophilus ATCC4356 and L. rhamnosus PTCC1607Inhibited bacterial growth and adhesion; stimulated IFN-G.
Yarmohammadi et al. (2021) [17]PreclinicalL. gasseri ATCC 33323Downregulated the expression of IL-8 and Bcl2.
Lin et al. (2020) [18]PreclinicalL. fermentum P2 (P2), L. casei L21 (L21), L. rhamnosus JB3 (JB3), or a mixture including the aforementioned three (multi-LAB) for 3 daysModulated metabolites important in immune response.
Maleki-Kakelar et al. (2020) [19]PreclinicalL. plantarumIncreased cell apoptosis.
He et al. (2022) [20]PreclinicalL. salivarius and L. rhamnosusAnti-inflammation (downregulated proinflammatory signaling pathways that included NF-κB, TNF, and IL-17; increased the abundance of beneficial bacteria in gut microbiota.
Lai et al. (2022) [21]PreclinicalParabacteroides goldsteinii MTS01Downregulated inflammation through downregulation of COX-2, IL-1β, and TNF-α; decreased pathogenic effect of H. pylori virulence factors.
Table 2. List of vaccine candidates against H. pylori and their potent immune response in animal models.
Table 2. List of vaccine candidates against H. pylori and their potent immune response in animal models.
AuthorsName of VaccineType of VaccineVaccine DeliveryTargetModel UsedType of Immune Response Elicited
Cen et al. (2021) [52]Saccharomyces cerevisiae-based oral vaccine EBY100/pYD1-UreB, EBY100/pYD1-VacA, or EBY100/pYD1-UreB + EBY100/pYD1-VacAVector-basedOralUrease and VacAMiceHumoral and mucosal immune response.
Wang et al. (2021) [53]L. monocytogenes-based vaccine, a multi-epitope chimeric antigen (MECU) containing multiple B cell epitopesVector-basedOral5 B-cell epitopes from FlaA, AlpB, SabA, and HpaAMiceElicited high levels of IFN-γ, IL-4, and IL-17 in splenic lymphocytes; increased IgA and IgG.
Xie et al. (2021) [54]Oral multivalent epitope vaccineMultivalent epitopeOralThree Th cell epitopes and five against B cellsMiceIncreased IFN-γ, IL-4, and IL-17 in lymphocyte supernatants to activate Th1, Th2, and Th17 mixed T-cell immune responses; increased IgA and IgG.
Liu et al. (2019) [55]Outer-membrane vesicles (OMVs) derived from gerbil-adapted H. pylori strain 7.13Outer membrane vesicleOralMembrane proteins of H. pyloriMiceTh2-biased immunity; increased IgA and IgG.
Peng et al. (2018) [56]Neutrophil-activating protein A subunit (NapA) and L. lactis as vectorVector-basedOralNeutrophil-activating protein A subunitMicePolarized Th17 and Th1 responses; increased IgA and IgG.
Espinosa-Ramos et al. (2019) [57]H. pylori 50–52 kDa immunogen-derived peptide antigen with the sequence Met–Val–Thr–Leu–Ile–Asn–Asn–Glu (MVTLINNE)Peptide antigenSubcutaneousImmunogen synthetic peptideMiceInduced thymus lymphocytes and significantly induced IL-6.
Pan et al. (2018) [58]Multivalent epitope-based vaccine cholera toxin B subunit (CTB)-HUUC with the intramucosal adjuvant CTB and tandem copies of B-cell epitopesMultivalent epitopeOral3 B-cell and 9 T-cell epitopesMiceH. pylori-specific lymphocyte responses and a mixed CD4+ T-cell immune response; increased IgA and IgG.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sukri, A.; Hanafiah, A.; Patil, S.; Lopes, B.S. The Potential of Alternative Therapies and Vaccine Candidates against Helicobacter pylori. Pharmaceuticals 2023, 16, 552. https://doi.org/10.3390/ph16040552

AMA Style

Sukri A, Hanafiah A, Patil S, Lopes BS. The Potential of Alternative Therapies and Vaccine Candidates against Helicobacter pylori. Pharmaceuticals. 2023; 16(4):552. https://doi.org/10.3390/ph16040552

Chicago/Turabian Style

Sukri, Asif, Alfizah Hanafiah, Sandip Patil, and Bruno S. Lopes. 2023. "The Potential of Alternative Therapies and Vaccine Candidates against Helicobacter pylori" Pharmaceuticals 16, no. 4: 552. https://doi.org/10.3390/ph16040552

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop