Next Article in Journal
In Vitro Analyses of Spinach-Derived Opioid Peptides, Rubiscolins: Receptor Selectivity and Intracellular Activities through G Protein- and β-Arrestin-Mediated Pathways
Next Article in Special Issue
An Analysis of Variability in the Content of Phenolic Acids and Flavonoids in Camelina Seeds Depending on Weather Conditions, Functional Form, and Genotypes
Previous Article in Journal
Probiotics Regulate Gut Microbiota: An Effective Method to Improve Immunity
Previous Article in Special Issue
Nanoformulations for Delivery of Pentacyclic Triterpenoids in Anticancer Therapies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Revisiting Therapeutic Strategies for H. pylori Treatment in the Context of Antibiotic Resistance: Focus on Alternative and Complementary Therapies

by
Ioana Alexandra Cardos
1,
Dana Carmen Zaha
2,*,
Rakesh K. Sindhu
3,* and
Simona Cavalu
2,*
1
Faculty of Medicine and Pharmacy, Doctoral School of Biomedical Sciences, University of Oradea, 1 University Street, 410087 Oradea, Romania
2
Department of Preclinical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, 1 University Street, 410087 Oradea, Romania
3
Chitkara College of Pharmacy, Chitkara University, Chandigarh 140401, India
*
Authors to whom correspondence should be addressed.
Submission received: 11 August 2021 / Revised: 1 October 2021 / Accepted: 2 October 2021 / Published: 8 October 2021

Abstract

:
The prevalence of Helicobacter pylori infection remains significant worldwide and it depends on many factors: gender, age, socio-economic status, geographic area, diet, and lifestyle. All successful infectious diseases treatments use antibiotic-susceptibility testing, but this strategy is not currently practical for H. pylori and the usual cure rates of H. pylori are lower than other bacterial infections. Actually, there is no treatment that ensures complete eradication of this pathogen. In the context of an alarming increase in resistance to antibiotics (especially to clarithromycin and metronidazole), alternative and complementary options and strategies are taken into consideration. As the success of antibacterial therapy depends not only on the susceptibility to given drugs, but also on the specific doses, formulations, use of adjuvants, treatment duration, and reinfection rates, this review discusses the current therapies for H. pylori treatment along with their advantages and limitations. As an alternative option, this work offers an extensively referenced approach on natural medicines against H. pylori, including the significance of nanotechnology in developing new strategies for treatment of H. pylori infection.

1. Introduction

The Helicobacter pylori is a helix-shaped, microaerophilic, Gram-negative bacterium, with high motility owing to its four to six flagella, capable of colonizing the gastric mucosa and thereby establishing infection. Most infected subjects remain asymptomatic for a long time, but this colonization produces a silent destruction of the gastric mucosa. Since its discovery in 1982, by Australian doctors Barry Marshall and Robin Warren, more and more studies have proven that H. pylori is responsible for atrophic gastritis, peptic ulcer, and gastric malignancies [1,2]. From a physiological point of view, H. pylori is microaerophilic, requiring oxygen to survive, but at a lower concentration than in the atmosphere. Another special feature of this bacteria is its nitrogen metabolism. As amino acids and urea are well known major sources of nitrogen in the gastric environment, the main necessary amino acids for H. pylori are valine, methionine, arginine, histidine, isoleucine, leucine, phenylalanine, fermentation being the most important mode of amino acid utilization [3]. On the other hand, all H. pylori strains produce high amount of urease (which accounts for up to 6% of total bacterial protein) as one of its adaptation methods to overcome stomach acidity. The mechanism involves the hydrolysis reaction of urea which results in ammonia and carbonic acid, and hence, neutralizes the strong acid in its environment, due to their buffering properties.
The arginase of H. pylori also plays an important role for acid protection in vitro, as the only pathway of arginine catabolism in H. pylori is that initiated by arginase [4,5]. In addition, another protection mechanism is involved, chemotaxis, H. pylori being able to detect the pH modifications in the gastric mucosa and move towards the less acidic region [6].
The H. pylori genome has been completely sequenced in 1980 being isolated from a patient suffering from gastritis [7]. It has a circular chromosome of 1,667,867 base pairs and 1590 predicted coding sequences [8]. The results suggest that the basic mechanisms of replication, cell division and secretion are similar to those of E. coli and H. influenza [8]. However, the size of its core genome (namely the number of genes present in isolates) varies depending on the number of analyzed strains and their geographical origin. Also, based on genome investigation, the virulence of H. pylori species has been related to their ability to produce a cytotoxin-associated protein (CagA) and active vacuolating cytotoxin (VacA) [9]. VacA is known to damage epithelial cells, disrupts tight junctions, and causes apoptosis, while CagA may cause inflammation and is a potentially carcinogen agent [10].
Actually, due to existing diversity between strains of H. pylori, the specific strain that infects a person can predict the outcome. On the other hand, the clinical aspects and the epidemiology of H. pylori infections have been drastically changing in the last decades, especially in developing countries. Despite the treatment regimens and international guidelines developed by specialists in microbiology, there is currently a noticeable failure, mainly due to H. pylori resistance to several antibiotics, in the context of global increase in antibiotic consumption in the general population [11,12]. For example, WHO admitted that clarithromycin-resistant strains of H. pylori are among the twelve priority pathogens for which novel antibiotics or alternatives are urgently required [11,13]. Many efforts have been made in order to understand the mechanisms of antibiotic resistance and hence, the sound understanding of its genetics and physiology attracted a huge interest form both fundamental and applied microbiology researchers, along with ecology, medical, and veterinary disciplines [14,15,16].
In the context of increasing rates of antibiotic-resistant H. pylori strains, the risk factors and prevalence on global population, the aim of our work is to highlight the main drawbacks of currently used treatment regimens against H. pylori and at the same time, to emphasize the huge potential of natural alternatives, plants extracts and new formulation design and strategies to combat this pathogen. Special attention is also given to nanotechnological formulations, with huge potential for tissue microenvironment-responsive treatment.

2. Prevalence and Risk Factors Relevant for Therapeutical Support

2.1. Prevalence of H. pylori Related to Different Geographic Areas and Global Population

The prevalence of H. pylori infections varies worldwide in terms of epidemiological, demographic and socioeconomic criteria. A systematic review with meta-analysis from 73 countries, in six continents, conducted by Zamani et al. for the period 1 January 2000–31 June 2017, revealed an overall prevalence of 44.3% worldwide and it is suggested that approximately 60.3% of the world population was infected in 2015 [17,18].
In terms of geographical distribution, Latin America and the Caribbean have shown the highest rate of infection in the world (59.3%), while at the opposite pole, in the Northern America, the prevalence was only 25.8%. While in North and South America, the pooled data presented a good homogeneity, large variations in country-based prevalence were noticed in Asia and Europe. For example, a high rate was observed in the population of Kazakhstan (79.5%) compared to those from Indonesia (10.0%), while in European countries, Serbia seems to have the highest prevalence (88.3%) in opposition to Belgium (11.0%) [19]. As expected, in terms of demographic and socio-economic criteria, in developing countries the rates were higher compared to the developed ones (50.8% vs. 34.7%), which can be related to the lifestyle and environmental parameters, fact agreed by another meta-analysis reported by K.Y. Hooi et al. [18].
Although the risk of exposure increases with the age, some studies suggest that children who became infected with H. pylori, were infected at very young age (<5 years), and approximately 85% have long asymptomatic periods and clinical manifestations are non-specific [20,21]. Another study from 2006 in Ireland concluded that most children become infected with H. pylori at a very young age, and the risk of infection declines rapidly after 5 years of age [20]. Obviously, children living in the most crowded conditions are at the greatest risk for H. pylori acquisition. An inverse correlation was noticed between the mother’s education and H. pylori positivity in children. On the other hand, it is generally accepted that breast-feeding plays a protective role against the acquisition of H. pylori infection [22,23]. However, in children, the treatment should be considered in a different approach than in adults, as the immune mechanisms are different.
In terms of epidemiology, many studies have proposed that acquisition of H. pylori occurs via common environmental sources such as drinking water [24,25], animals or animal products [26,27,28]. The most probable route of transmission seems to be the ingestion route [29].
Even if H. pylori-associated disorders usually regress or heal completely after successful treatment with antimicrobials, the available antimicrobial therapies for H. pylori infection have many shortcomings and side effects. A combined therapy is very often required, especially because of the development of antimicrobial resistance [30].

2.2. Risk Factors: Oncogenicity

H. pylori is extremely variable in prevalence, taking into consideration risk factors such as: age, gender, socio-economic status, geographic area, and diet and lifestyle. It is widely accepted that lower socio-economic status is linked to poor hygiene conditions and lack of sanitary education, thus leading to higher rates of H. pylori incidence. That is why the prevalence of H. pylori infection is higher in developing countries and lower in high-industrialized countries. Beside the socio-economic status, deficient sanitation, and crowded living conditions, which are well known risk factors, some studies suggested that smoking, use of nonsteroidal anti-inflammatory drugs, 0 blood group, high body mass index and family history of gastric disease are also considered risk factors in the acquisition and transmission of H. pylori [31,32,33]. Diet may be another risk factor for H. pylori infection. Several epidemiological studies have revealed that good dietary habits such as consumption of green tea, fruits, vegetables with high levels of carotenoids, folate, vitamin C and phytochemicals are protective against H. pylori infection and gastric cancer risk [34,35,36,37,38].
Salt and some transition metals such as iron, zinc, nickel, can alter H. pylori virulence and accelerate H. pylori-dependent disease progression [34]. H. pylori can also exert its carcinogenesis effect directly over its virulence factors such as cytotoxin-associated gene A (CagA) containing the Cag Pathogenicity Island (CagPAI), vacuolating cytotoxin A (VacA). The strains protecting CagA can cause more inflammatory responses (blood group antigen-binding adhesin A (BabA) and progression to gastric cancer. CagPAI encodes a bacterial type IV secretion system which secretes CagA and peptidoglycan into the epithelial cells. CagA and peptidoglycan induce multiple cellular signaling pathways such as MAPK cascade and phosphoinositide-3 kinase (PI3K-AKT) signaling pathways, respectively, as described in Figure 1. The explanation is based on the fact that the most important virulence factors expressed by H. pylori, VacA and CagA toxins, are transcriptionally regulated in by iron [39].
On the other hand, several studies have shown a correlation between zinc intake and H. pylori-dependent disease progression in the Asian population, which are well known related to CagA strains of H. pylori [34,40,41,42]. There are also differences between genders and age. A systematic review and meta-analysis published in 2017 showed that male gender is associated with a higher prevalence rate for H. pylori than the female gender, both in grown-ups and in children [43]. In a study from 2003 that included 307 children and 604 adults, H. pylori was diagnosed in 40 children and 308 adults [44]. There is also evidence that it plays a significant role in gastric cancer [45,46,47,48,49,50]. Gastric cancer is the number 5th most frequently diagnosed malignancy worldwide. It has a poor life expectancy despite medical treatment, being the 3rd cause of death due to cancers worldwide [45]. H. pylori has been recognized as a first-class risk factor for gastric malignancies by the International Agency of Cancer Research since 1994. Based on histological characteristics, gastric cancers can be classified in two major types: adenocarcinoma and lymphoma of gastric mucosal-associated lymphoid tissue (MALT). According to several scientific data, the second one is less common than adenocarcinoma (only 3% of all gastric tumors), while MALT-lymphoma is in 90% of cases caused by H. pylori infection [46,47]. Effective eradication of H. pylori has been proved to improve clinical outcome of patients suffering from gastric ulcers, preventing clinical relapses, lowering the risk of MALT-lymphoma incidence and inducing regression in diagnosed MALT-lymphomas [51,52,53]. Therefore, it is of crucial importance to evaluate the time frame in which H. pylori causes a carcinogenic effect, because beyond this time point, the intervention is significantly less effective. Taking into account the virulence factors and metabolic processes of H. pylori in developing gastric carcinogenesis, a complex approach should be consider including multiple measures and usage of appropriate antibiotics, eventually combined with natural biomedicines are necessary in order to eradicate this bacterium.

3. Current Treatment Regimens for H. pylori Infections

H. pylori seen as a bacterial infection, has been intensely studied, concerning a proper treatment. Although in vitro testing made it seem sensitive to a wide range of antimicrobials, in vivo only a few are useful [54]. Even so, there are more factors that must be taken into consideration such as: doses, formulations, frequency of dosing in relation to meals, treatment duration, use of anti-secretory drugs, antiacids, or probiotics [55]. The success of the therapy for many antimicrobial drugs is greatly diminished by special conditions such as acidic pH, formation of a biofilm but antibiotic resistance is the most important cause of failure of H. pylori eradication. The mechanisms underlying antimicrobial resistance are well known and they are also applicable for H. pylori. There are only a few antibiotics active against H. pylori: clarithromycin, amoxicillin, metronidazole, fluoroquinolone (levofloxacin, norfloxacin) tetracycline, rifabutin. Of concern is the increase in resistance to clarithromycin and metronidazole. It is currently accepted that H. pylori effective therapy requires two or more antimicrobials for a period of 14 days, along with an antisecretory drug to reduce gastric secretion.
The best treatment antibiotic option is established by drug-susceptibility testing which identify susceptible and resistant strains. There are many methods for this purpose, Kirby Bauer method, microdilution, E-test but all are time consuming and not available in all laboratories. Instead, a better option are molecular methods that detect specific changes in the H. pylori genome [56,57,58].

3.1. Clarithromycin Triple Therapy

International guidelines for treating infection with H. pylori are consistent with the use of triple therapy as the first-line treatment. Triple therapy consists of the administration of a proton pump inhibitor (PPI), clarithromycin, and amoxicillin for one-two weeks [59,60]. Currently, concomitant therapy (PPI, amoxicillin, clarithromycin, and a nitroimidazole administered concurrently) should be the preferred non-bismuth quadruple therapy, as it has shown to be the most effective to overcome antibiotic resistance according to the Maastricht V Convention [59]. The clarithromycin regimen is one of the first and most effective eradication therapies. The regimen has a high-eradication rate >95%, but the best results are obtained if the regimen is followed for 14 days [61,62]. For various reasons, sometimes is prescribed for 7 or 10 days, leading to lower rates of eradication mainly due to an increase in clarithromycin resistance [63]. Other regimens called second-line therapies have been proposed and applied [63]. These treatments consist of an association of PPI with two or three antibiotics: amoxicillin, clarithromycin, metronidazole. Lately, it was proven that rates of eradication go as far as 70%, which is considered very low for an infectious disease. Concomitant therapy is preferred over sequential one because it is easier to be administrated. There are controversies about superiority of concomitant therapy toward sequential therapy [64,65,66,67], but there is relevant evidence proving that there is no significant difference in the eradication rate between concomitant therapy and sequential therapy [68,69]. In case of resistance to both metronidazole and clarithromycin, the alternative candidates for third-line therapy are quinolones, tetracycline, rifabutin, and furazolidone, but the rate of resistance to metronidazole and quinolones used as second- or third-line regimens is more than 15% worldwide [70,71,72].

3.2. Bismuth Quadruple Therapy. Non-Bismuth-Based Quadruple Therapy

The bismuth quadruple therapy consists of administrating bismuth, proton-pump inhibitors, tetracycline, metronidazole or tinidazole for a period of 7–14 days. In the 19th century bismuth was widely used in gastroenterology. Since 1970, bismuth was used as colloidal bismuth subcitrate (De-Nol), as an ulcer medication. It has many pharmaceutical properties such as: bactericidal effect on H. pylori, binding to the ulcer base, inactivation of pepsin, binding of bile acids, stimulation of prostaglandin biosynthesis, suppression of leukotriene biosynthesis, stimulation of complexation with mucus, inhibition of various enzymes, binding of epithelial growth factor, stimulation of lateral epithelial growth [73]. The bismuth quadruple therapy has an excellent H. pylori eradication rate (more 80%) therefore is considered a second line therapy [74,75,76]. Bismuth was used as a medical agent ever since 1697 [76]. Its disadvantages are the inaccessibility to bismuth in some countries, the rising resistance to metronidazole and low adherence to the regimen [77]. A single capsule therapy containing bismuth, metronidazole, and tetracycline has been developed and administrated with large successful rates [78]. According to a meta-analysis, first- and second-line single-capsule therapy as described above achieved an eradication rate approaching 90% [79]. The non-bismuth-based quadruple therapy consists of adding metronidazole to the classic PPI, amoxicillin, and clarithromycin. It is efficient in clarithromycin resistant populations, but it has not been studied for metronidazole resistant populations. Eradication rates can go as high as 88%. It is safe to use, efficient, but it has gastro-intestinal side-effects [80,81].

3.3. Rifabutin-Based Triple Therapy

Rifabutin is a structurally similar to rifampicin use as anti-tuberculosis drug, mainly for atypical tuberculosis. The action of rifabutin consists in stopping protein synthesis by inhibition of the beta-subunit of the Helicobacter DNA-dependent RNA polymerase. This enzyme is encoded by RpoB. In case of mutation of RpoB results resistance to rifabutin [82]. The rifabutin-based triple therapy used as a third-line therapy in patients that failed to response to classic antibiotic treatments such as amoxicillin, clarithromycin, metronidazole, tetracycline, and levofloxacin. It consists of rifabutin, amoxicillin and a PPI taken for a period of 14 days. The ideal dosage is 150–300 mg rifabutin daily. The therapy with rifabutin has many advantages (fat-soluble, good absorption after oral intake, stable at a wide range of pH values), but the most important is low the rate of resistance in H. pylori because it is rarely used clinically [83]. Rifabutin-based regimens are important for rescue therapies, but the eradication rate is different reported and future related studies are needed. Overall, the efficacy rates of eradication are 73%. Its disadvantages include leucopenia and myelotoxicity [84,85].

3.4. Fluoroquinolone Therapy

The levofloxacin triple therapy is used because of increasing resistance to clarithromycin. The ideal dose is 2 mg × 500 mg of levofloxacin daily, with fewer side-effects than the bismuth quadruple therapy. It is used as a second-line rescue therapy, after first-line eradication treatments fail [86]. New generations of fluoroquinolones are levofloxacin, moxifloxacin, gatifloxacine, and pefloxacine. They have a broad-spectrum activity and are used in infections of the respiratory and urogenital tract. Because they are more frequently used, the bacterial resistance, including H. pylori, is rising [87]. Data containing other fluoroquinolones, other than levofloxacin, are rare. Moxifloxacin used with esomeprazole and tinidazole or moxifloxacin with esomeprazole and amoxicillin had eradication rates of about 88–90% in a study from 2005 [88,89]. A study from India showed an eradication success of up to 77% for a 2-week dual therapy with proton pump inhibitors and norfloxacin [90,91]. They are used as a second line therapy in countries where bismuth is not available or where the population has high rated of clarithromycin resistance.

3.5. Potassium-Competitive Acid Blockers (P-CABs)

Potassium-competitive acid blockers are a group of drugs that inhibit gastric secretion faster and with a longer action. They seem to be more potent than PPI because they do not require activation by stomach acid and their efficacy is not influenced by various subtypes of cytochromes P450. The P-CABs are bases that inhibits the H+ K+ ATPase enzyme, key enzyme in formation of HCL. In addition, the protonated form of this drug is less able to pass via cellular membranes than the nonionic molecules, so they concentrate into the canaliculi of parietal cells, the place where HCl is produced [92]. The efficacy for H. pylori eradication was assessed in more multicenter studies using P-CABs in association with antibiotics (triple therapy). The eradication rate was 94.4% for the first-line therapy and 97.1% for the second-line therapy. The conclusion was that P-CABs-triple therapy was safe and effective [79], while the eradication and the incidence of adverse events was 4.4% [93].

4. Antibiotic Resistance and Alternative Therapy

The 1980 discovered H. pylori bacteria is nowadays considered as the etiologic agent for gastric ulcers and gastric cancer. Earlier, various drug regimens were used for the treatment and cure of such bacterial infections. However, now, the alarming situation of increasing resistance to antibiotics, moreover the high-cost therapy, resulted into the emergence of alternative approaches such as anti-inflammatory agents, anti-oxidants and most importantly, herbal plants and their extracts. These alternative approaches inhibit and modulate the release of inflammatory cytokines, reactive oxygen species and other inflammatory mediators which generate on the interaction of H. pylori with gastric mucosa and ultimately lead to atrophy, dysplasia, metaplasia and changes in the gastric gland. The herbal extracts have shown important roles such as anti-inflammatory, antioxidant, anti-cancer, and bactericidal against H. pylori [94,95]. There are alternative therapies for the treatment H. pylori infection as shown in Figure 2.
As a result, there is an increasing trend of herbal drug therapy in almost 80% of population, especially those who found modern antibiotic therapy not affordable, not accessible, and not accepted. Various plants and their extracts show promising results against H. pylori [94]. A selection of the most relevant natural extracts was performed by searching the full text information in databases of PubMed, Web of Science, Embase, and Google Scholar, with priority within the time interval 2005–present, as presented in Table 1.
In the next section, a more detailed discussion is provided, referring to propolis, curcumin, ginseng, and garlic extracts, as a valuable source of active ingredients against H. pylori mediated disease, due to their easy accessibility and affordability for population.

4.1. Role of Propolis in Treatment of H. pylori Mediated GIT Diseases

One of the important and beneficial natural treatments for H. pylori infection is propolis, produced by honeybee Apis melifera. Different studies conducted all over the world reported that caffeic acids, flavonoids, and phenolic esters are the main biologically active compounds in propolis [126,127]. Specifically named chemical compounds (to mention only a few) arechalcones-2,6-dihydroxy-4-methoxychalcone and 2,4,6-trihydroxy-4-methoxy-chalcone, benzoic acid derivatives such as salicylic acid, 4-methoxybenzoic acid, 4-hydroxybenzoic acid, procatechuic acid, gallic acid, etc., benzaldehyde derivatives such as vanillin and isovanillin, cinnamyl alcohol, cinnamic acid and its derivatives, terpene, sesquiterpene alcohol and their derivatives, aliphatic hydrocarbons, sterols and steroid hydrocarbons, sugars such as d-glucose, d-fructose and ribofuranose, amino acids, and some minerals such as Na, K, Mg, Zn, Cd, Ca, Ag, Ni, Fe. These mentioned compounds and their derivatives account for the pharmacological importance of propolis [128].
The bioactive phenolic compounds are present as free or alkyl and phenyl esters which are responsible for various antioxidant, antimicrobial, anticancer and anti-inflammatory properties of propolis. According to literature, 21 polyphenols were detected in propolis which account for the anti H. pylori activity [128,129,130,131]. The main compounds and their combination of chrysin, pinocembrin, galangin and caffeic acid phenethyl ester showed the bactericidal effect against H. pylori. The research studies conducted by Shapla et al. [128] have proven the scientific evidence in treatment of H. pylori infection and clearly justifies the protective role of propolis phenolic compounds in decreasing and neutralizing the host and bacteria’s physiology. Hence, they suggest propolis as an alternative treatment approach in H. pylori associated gastrointestinal diseases [128].

4.1.1. Anti-Inflammatory Effect of Propolis

The caffeic acid phenethyl ester (CAPE) obtained from propolis also has anti-inflammatory properties. It shows immunosuppressive action on T cells of host and also inhibits cell activation cycle. It also shows inhibitory effect on other inflammatory mediators such as NFkB and transforming growth factor. In an in vitro study CAPE blocks the MAPK signaling and NFkB activation and ultimately decrease IL-8 production, because IL-8 production is an important pathological role in H. pylori induced gastritis. In addition to the inhibitory effect on mediators, it also shows its action on enzymes such as myeloperoxidase, and NADPH-oxidase, and this activity indirectly inhibits neutrophils and other inflammatory cells infiltration. The other important enzyme peptide deformylase in pathogenesis is also competitively inhibited by CAPE. The oxygen free radicals produced by Xanthine oxidase also cause tissue damage and inflammation which are inhibited by propolis extract [132,133].

4.1.2. Antioxidant Effect of Propolis

Through research studies propolis is found to have higher antioxidant effects than green tea. It contains various polyphenols and flavonoids which show antioxidant properties in many ways such as hydrogen donors, reducing agents, chelating ions, etc. It protects the blood cells from oxidative stress induced by free radicals and maintain their cellular structures, rigidity, and their functions [126,133,134]. Hence, based on several evidence presented, the role and benefits of propolis are clearly against H. pylori infection. Therefore, it can be concluded that propolis may be used as an alternative therapeutic approach in the treatment and ailment of the gastrointestinal diseases such as ulcers and gastritis.

4.2. Role of Curcumin in Treatment of H. pylori Mediated GIT Diseases

Curcumin is a well-known polyphenolic bioactive yellow color pigment obtained from rhizomes of curcuma longa also known as turmeric. It is widely used as a spice and food coloring agent especially in India. Various research studies previously shown multiple health benefits such as antioxidant, anti-inflammatory, anti-cancer, and anti-microbial, with the approach of anti-angiogenesis for the prevention and treatment of certain diseases such as cancer, chronic inflammation, or atherosclerosis [135]. The invitro studies and preclinical trials also gave results on its effect on H. pylori and its induced diseases. From a chemical point of view, curcumin exists as a keto-enol tautomer, where the enol isomer is more stable than keto form, in both solid and liquid state. The yellow color is found at acidic pH, while basic pH shows red color of curcumin. The polyphenolic chemical compounds possess two aromatic rings with two unsaturated carbonyl groups. This molecule is stable due to the central hydroxyl group (-OH) which is found to be the crucial functional group for antioxidant and other biological activities [136].

4.2.1. Antioxidant Effect of Curcumin

The curcuminoids in alcoholic extract reported to act as scavengers for oxygen free radicals. They interact with reactive oxygen intermediates and protect the cells from damage. Curcumin is found to be more stable at acidic pH in GIT. Oxidative stress and damage play a vital role in pathogenesis of H. pylori induced diseases. So, it has shown to be a potent oxidative scavenger to treat oxidative induced diseases [137].

4.2.2. Anti-Inflammatory Effect of Curcumin

The anti-inflammatory effect of curcumin is already well known. Curcumin inhibits carrageenan- induced paw edema in mice in various inflammatory diseases such as ulcerative colitis in preclinical trials. The mechanism behind the anti-inflammatory effect is inhibition of nitric oxide synthase, cyclooxygenase-2, and inflammatory mediators such as cytokines (IL-1, IL-6, IL-8, IL-12), tumor necrosis factor-α etc. It also inhibits prostaglandins along with lipoxygenase activity [138].

4.2.3. Anti-Carcinogenic Effect of Curcumin

Curcumin has also anti-cancer property and plays an important role in treatment of gastric and other types of cancer. The inflammation and malignancy are correlated. The inhibition of I-Kβ kinase, and various transcription factors reduces the effect of inflammatory mediator NF-Kβ and synthesis of cytokines, respectively, which is involved in pathogenesis of cancer. Along with gastric cancer treatment it is also used in oral, esophageal, duodenal, and colon cancer [139].

4.2.4. Anti-Microbial Effect of Curcumin

Curcumin inhibits the growth of various bacteria’s and parasites including H. pylori, Bacillus subtilis, Plasmodium falciparum, Leishmania, etc. Curcumin, the Indian gold has all the properties from being an antioxidant to a beneficial anti-inflammatory agent, that is why it is used in treatment of H. pylori bacterial infection. The invitro studies by Marathe et al., gave results on inhibition of growth of 19 different strains of H. pylori [140]. Using a mouse model, Vetvicka et al. [141] studied the effects of curcumin on lipid peroxide level, myeloperoxidase and urease activity, along with levels of anti- H. pylori antibodies, biofilm formation, and levels of IFN-γ and IL-4 in serum. They found significantly increased IL-4 and decreased IFN-γ serum levels, while the levels of LPO (an oxidative damage index) were decreased by supplementation with curcumin. The results of this study also support the hypothesis that curcumin has strong immunostimulating properties.
Hence, curcumin can be used either alone or in combination with other antibiotics to cure and treat H. pylori induced gastric diseases due to its multiple benefits.

4.3. Role of Ginseng in Treatment of H. pylori Mediated GIT Diseases

Since centuries, Ginseng, obtained from the roots of Panax ginseng is a widely used herb in Asian folk medicine. The antioxidant, anti-inflammatory, anti-microbial, and anti-cancer properties of ginseng have already been proved in many pharmacological and clinical studies [142,143,144]. Ginseng mainly contains saponins called ginsenosides, which are secondary metabolites, responsible for pharmacological activity. Actually, the structural diversity of ginsenosides contributes to the multiple pharmacological benefits of ginseng on cancer and diabetes treatment, inflammation, stress, immune, cardiovascular system, and central nervous system disease.
The acidic polysaccharide from ginseng helps to dislodge the H. pylori from human gastric epithelial cells and Ginsenoside Rb1, an important primary Ginsenoside improves inflammation by inhibiting pro-inflammatory mediators mainly cytokines and TNF-α.
Ginseng inhibits DNA mutagenesis, pathogen mediated hemagglutination, improves immune–modulatory functions and exerts bactericidal effect and protects the host from pathogen induce inflammation. The lactic acid present in fermented ginseng extract also exhibits potent anti- H. pylori activity [144].
Another research study shows the beneficial effect of red ginseng as it exerts protective function against carcinogenesis in gastric mucosal cells [143]. Red ginseng inhibits ERKI/2 signaling which on further inhibits DNA damage and apoptosis induced by H. pylori. It also inhibits synthesis of pro-inflammatory mediators especially IL-1β, iNOS, MOP activity. Panaxytriol shows effective inhibition of H. pylori growth and Ginsenoside Rh1 and protopanaxatriols inhibits the terminal stage H+/K+ ATPase of stomach acid secretion [145,146].
White ginseng extract has also shown antimicrobial, anticancer, and anti-inflammatory effects. Through disk diffusion assay study, it has been concluded that it inhibits the bacterial growth effectively. Additionally, it exerts cytotoxic effects and used in treatment of lung cancer, endometrial and uterine carcinoma. Moreover, the anti-inflammatory effect is shown by inhibiting nitric oxide production and activity [144,145]. In a very recent paper, [147] it was demonstrated the inhibitory effect of Korean red ginseng extract on DNA damage response and apoptosis in Helicobacter pylori-infected gastric epithelial cells, by increasing apoptotic indices and ROS levels. The authors suggests that supplementation with Korean red ginseng is beneficial for preventing the oxidative stress-mediated gastric disorders associated with H. pylori infection.
Therefore, it can be concluded that ginseng may be used as an alternative or complementary therapeutic approach in the treatment of H. pylori induced infections.

4.4. Role of Garlic in Treatment of H. pylori Mediated GIT Diseases

Currently, the principal medicinal uses of garlic are to prevent and treat cardiovascular disease by lowering blood pressure and cholesterol, as an antimicrobial agent, or as a preventive agent against different types of cancer. The antimicrobial activity of allicin, a diallyl thiosulfinate compound, was identified in garlic by Cavallito and Bailey in 1944 [148]. The garlic extract was found to be effective against H. pylori infection, as thiosulfinates (allicin) play an important role in the antibiotic activity of garlic [96]. Based on a systematic review and meta-analysis, Xiao-Bei Si et al. [98] highlighted the output of clinical trials that included allicin as an add-on treatment to triple therapy or bismuth containing quadruple therapy for H. pylori infection. The results of this meta-analysis revealed that allicin treated groups showed an eradication rate of ~93.81%. Additionally, higher healing rates and total remission rates of peptic ulcers were evidenced, and different scenarios for the mechanisms involved in ulcer response were proposed: (i) the activation of NF-Kβ was inhibited by allicin, which further inhibits the production of TNF-α, leading to anti-inflammatory effects; (ii) garlic extracts possess a protective role against oxidative stress in gastric tissue, which cause mucosal injury and ulcer development; (iii) garlic extracts favors NO synthesis, increasing the activity of nitric oxide synthase with crucial role in endothelial function [98].
The Feldberg in vitro research study has elucidated the mechanism behind the antimicrobial activity [144]. Allicin mainly inhibits the mRNA degradation as well as RNA, DNA and protein synthesis in the cells, where RNA polymerase is the major target of allicin. The major garlic materials available are garlic powder and garlic oil. Garlic powder is prepared by slicing, drying, and pulverization of garlic cloves, whereas the oil is prepared by heating the cloves and collection of its vapor. These commercial formulations are widely used for its antimicrobial activity against H. pylori [97]. Chemopreventive activity of garlic was also demonstrated either as fresh extract, garlic oil, or organosulfur compounds derived from garlic [149,150,151]. Garlic is also rich in antioxidants which destroys free radicals that can damage the cell membrane of epithelial cells, inducing gastric cancer. The antioxidant activity of garlic is mainly due to the organosulfur compounds derivatives which inhibit the pathological mechanisms involved in cancer, including free radicals formation, mutagenesis, angiogenesis, DNA formation, and cell proliferation. The cell proliferation is inhibited by garlic, mainly acting on G2/M phase of cell cycle [96,152].
With the evidence provided by the studies, it is clear that garlic serves as beneficial alternative therapy to antibiotics against different gastrointestinal tract diseases induced by H. pylori.

5. Nanotechnology-Based Approach against H. pylori Infections

Metallic NPs such as silver, gold, zinc or iron have been previously reported to possess the ability of killing a wide range of bacteria including H. pylori [153,154] by well-known underlying mechanisms involving oxidative stress, metal ion release and nonoxidative stress. A very low NPs concentration is necessary for bactericidal effect, and hence, it is difficult for the bacteria to develop resistance. Among different metallic NPs, AgNPs are convenient, especially the biologically derived ones, as the preparation methods demonstrated a controlled particle size, shape, and mono-dispersity, while reducing time of preparation, in the context of environmentally friendly approaches.

5.1. Role of Biologically Synthesized Silver Nanoparticles

Biosynthesis of silver nanoparticles using different plants extracts, along with other biological sources such as bacteria, fungi, algae, emerges as a good alternative production, using nontoxic, clean and eco-friendly approach, allowing to modulate its physicochemical parameters. The concentration of plant extract and the substrate, reaction temperature and pH strongly affect size, shape and stability of silver nanoparticles. The number of publications dealing with AgNP synthesis with the assistance of various plant extracts (leaves, stems, roots, etc.) is huge, but recently, several papers aiming to highlight the importance of biosynthesized AgNPs, demonstrated that the main advantage of AgNPs synthesized from plant extract is that they are more stable compared to microorganisms synthesized nanoparticles [155,156]. The study conducted by Gurunathan et al. [157], AgNPs were biologically synthesized using leaf extract of Artemisia princeps as a bio-reductant and evaluated against H. pylori. The average size distribution of the synthesized particle was between 10–40 nm. The main results of this study demonstrated a dose-dependent decrease in cell viability and biofilm of H. pylori formation, potentiation of reactive oxygen species generation and DNA fragmentation in H. pylori. Moreover, cytotoxic effect against human carcinoma cells was evidenced [157]. According to literature [158], the mechanism of AgNPs action on cells involve several steps: adhesion on the surface of the bacterial cell wall (especially observed for Gram-negative bacteria, knowing to have a thicker cell wall), penetration into the cell and disruption of intracellular organelles and biomolecules, induction of oxidative stress, and modulation of signal transduction pathways. It is generally accepted that the bactericidal properties of AgNPs are associated with a disruption of RNA transcription, purines, pyrimidines, and fatty acids of bacteria, while upon interaction of AgNPs with cell DNA, the replication can break [158].
Muhammad Amin et al. [159] performed the green synthesis of silver nanoparticles using methanol extract from the plant Solanum xanthocarpum and analyzed its anti H. pylori activity in vitro, by comparing with four standard drugs: amoxicillin, clarithromycin, metronidazole, and tetracycline.
The results of this study indicated a stronger anti- H. pylori activity of AgNPs compared to AgNO3 and metronidazole, showing almost similar results to tetracycline, but less potent than amoxicillin and clarithromycin. Moreover, a significant inhibition was noticed in the H. pylori urease inhibitory assay. It was suggested that biological silver nanoparticles act as a potent therapeutic compound with anticancerous and antimicrobial properties, opening a new area of research for large medical applications of silver nanoparticles, including eradication of the H. pylori mediated gastric diseases [159]. Besides silver, other metallic nanoparticles such as bismuth, zinc, and gold were proved to be effective in the treatment of H. pylori, as reviewed by Safarov et al. [160]. Although there are enough evidences of the antimicrobial effect of metallic nanoparticles, there are also limitations, as most of them tend to accumulate in kidneys and liver, leading to cytotoxicity.

5.2. Other Nanoparticles Formulations for H. pylori Mediated GIT

5.2.1. Polymeric Nanoparticles Encapsulating Drugs and Other Therapeutic Agents

Polymer-based nanocarriers are advantageous formulations when it comes to gastric delivery as they exhibited very good stability and loading capacity. The drug or therapeutic molecules can be directly encapsulated with polymeric nanoparticles or covalently conjugated to their surface. The role of nanocarriers is to protect the bioactive compounds in harsh gastric environments, to enhance the internalization by microbes and eradicate them in multiple pathways.
PLGA (poly lactic- co- glycolic acid) are good candidates because due low toxicity and possibility to encapsulate low water-soluble drugs such as clarithromycin, in order to reach the target region [161,162]. PLGA nanoparticles loaded with clarithromycin, in size of about 360 nm, have shown MIC values in the range 0.003 µg/mL to 0.05 µg/mL, which were significantly lower than non-encapsulated drug solution, as demonstrated for clinical five H. pylori strains [162]. Gliadin, a biodegradable, biocompatible, and natural origin biopolymer, is known to adhere to mucosa membranes. Thus, its mucoadhesive properties is advantageous for the development of nanoparticulate delivery systems against H. pylori, as demonstrated by Ramteke S. et al. [163]. When using omeprazole and clarithromycin together, in a gliadin nanoformulation, the anti- H. pylori effect was found to be over 80%. Clarithromycin was also encapsulated into ethylcellulose nanoparticles and its activity against H. pylori was investigated by Pan-In, P. et al. [164] in animal model (rats).
The encapsulated clarithromycin was more effective than the free clarithromycin formulation, when administrated in rats. Actually, the initial reason for the antibiotic encapsulation was mainly due to its susceptibility to the highly acidified environment in the stomach.
Chitosan is another non-toxic polysaccharide, cationic in nature, with mucoadhesive properties, and widely used in the form of nanoparticles for gastric delivery of different bioactive compounds. It is composed of D-glucosamine and N-acetyl-d-glucosamine units, being insoluble at neutral pH, while rapidly ionizes and dissolves under the acidity of stomach [165]. The potential utility of chitosan micro and nanoparticles in the treatment of gastric infection was evaluated by Gonçalves et al. [166]. Enhanced effect of encapsulating amoxicillin into chitosan nanoparticles to treat H. pylori was demonstrated in vivo using Wistar rats, highlighting that H. pylori inhibition was not influenced by particle size [166]. Alginate, heparin, and poly-g-glutamic acid are anionic polymers commonly used for preparation of chitosan-based polyelectrolyte complexes, in the form of micro- or nanoparticles, and have been developed for encapsulation of antibiotics for H. pylori treatment [167,168]. According to literature, the novel strategy using targeted treatments based on chitosan micro/nanoparticles, specific to H. pylori, may include chemically modified (functionalized) particles with targeting compounds that bind H. pylori urease, flagella, adhesins, aiming to inhibit, kill or remove the bacteria, while avoid the destruction of the normal gut microbiome of the patients.

5.2.2. Liposomal Formulations against H. pylori

Liposomes are also widely studied drug-delivery systems for antibiotic delivery, consisting of spherically assembled phospholipid bilayers, encapsulating various antibiotics, including metronidazole, ampicillin, amoxicillin, and doxycycline against to H. pylori [169]. The encapsulation efficiency of these drugs was found to be relatively low, and the in vivo effect of these formulations against H. pylori has not been verified. Fatty acids such as stearic acid, oleic acid and linoleic acid have been encapsulated into liposomes and evaluated against the resistant strains of H. pylori [170]. The hydrodynamic diameter of these formulations was around 86 nm. Both the liposomal formulation and free linoleic acid were effective in killing spiral and coccoid forms of the bacteria. The results demonstrated that linoleic acid possess the most relevant bactericidal effect thanks to its increased permeability, being capable of completely eliminate eliminating H. pylori within 5 min at a concentration of 400 μg/mL [170]. Because of its amphiphilic nature, linoleic acid can be readily loaded into liposomes and subsequently fuse with bacteria for antibacterial activity. Moreover, in vivo tests (mice animal model) confirmed that linoleic acid-based liposomal formulation was efficient in killing H. pylori and reducing bacterial load in the mouse stomach [171]. Additionally, the treatment was efficient in reducing the levels of proinflammatory cytokines including interleukin 1β, interleukin 6, and tumor necrosis factor alpha.
Other liposomal formulations were prepared using incorporation of docosahexaenoic acid into nanostructured lipid carriers, which resulted in increased bactericidal efficacy at much lower concentration compared to free docosahexaenoic acid. The suggested mechanism was related to the fragmentation of H. pylori cell membrane upon interaction with nanoliposomes, and leakage of cytoplasmic content, as revealed by Transmission Electron Microscope images, acting very rapidly in a dose-dependent manner [172]. An interesting approach used an engineered liposomal formulation based on gold/chitosan nanoparticles adsorbed on the liposomes surface, which demonstrated a smart “on-demand” antibiotic delivery in a controlled manner: the more enzymes or bacteria present at the infection site, the more antibiotic molecules released in situ [173]. Such on-site release of antibiotics enables localize and rapid killing of H. pylori, and minimizes side effects.

6. Conclusions

H. pylori is responsible for a chronic, transmissible, infectious disease and the increasing prevalence of antibiotic resistance has complicated the therapy. All therapies should assume the possibility of antimicrobial drug resistance. As an infectious disease, therapy should be based on drug susceptibility testing, but it is not always available. Since its classification as a group 1 carcinogenic by International Agency for Research on Cancer, substantial effort has been allocated in the investigation of therapeutic alternatives beyond antibiotics. Clarithromycin triple therapies now typically produce less than 80% cure rates and thus is no longer acceptable. Bismuth and non-bismuth-based quadruple therapy are inaccessible in some countries, and often are accompanied by gastro-intestinal side-effects. The rate of eradication in the case of rifabutin-based triple therapy is about 73%, but its disadvantages include leucopenia and myelotoxicity, while fluoroquinolone therapy, as a second line therapy in countries where bismuth is not available or where the population has high rated of clarithromycin resistance, may offer eradication success of up to 77%. The P-CABs triple therapy is probably the most safe and effective against H. pylori, but the treatment may cause individual variability regarding the effect. Selection of appropriate treatment should be made taking into account the molecular mechanisms of pathogenesis and virulence factors engaged in the development of gastric disease, while keeping in mind that successful antibacterial therapies depend on susceptibility to given drugs, doses, formulations, use of adjuvants, treatment duration, and reinfection rates. In this respect, the use of natural compounds in H. pylori treatment has gained huge popularity due to their low side effects and low toxicity, as encouraging results from in vitro and in vivo studies were reported. This work offers an extensively referenced approach on natural medicines against H. pylori, including the significance of nanotechnology in developing new strategies for treatment of H. pylori infection.

Author Contributions

S.C., D.C.Z., I.A.C. and R.K.S. contributed to the reading and selection of the papers included in references list; R.K.S. performed the graphical interpretation. I.A.C., R.K.S., S.C. and D.C.Z. prepared the original draft; I.A.C., S.C. and D.C.Z. reviewed the final version of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kandulski, A.; Selgrad, M.; Malfertheiner, P. Helicobacter pylori infection: A clinical overview. Dig. Liver Dis. 2008, 40, 619–626. [Google Scholar] [CrossRef] [PubMed]
  2. Kim, S.Y.; Chung, J.W. Best Helicobacter pylori Eradication Strategy in the Era of Antibiotic Resistance. Antibiotics 2020, 9, 436. [Google Scholar] [CrossRef] [PubMed]
  3. De Reuse, H.; Skouloubris, S. Nitrogen Metabolism. In Helicobacter pylori: Physiology and Genetics; Mobley, H.L.T., Mendz, G.L., Hazell, S.L., Eds.; ASM Press: Washington, DC, USA, 2001; Chapter 11. Available online: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/books/NBK2412/ (accessed on 1 August 2021).
  4. Weeks, D.L.; Eskandari, S.; Scott, D.R.; Sachs, G. A H+-gated urea channel: The link between Helicobacter pylori urease and gastric colonization. Science 2000, 287, 482–485. [Google Scholar] [CrossRef] [Green Version]
  5. Quintero, M.J.; Muro-Pastor, A.M.; Herrero, A.; Flores, E. Arginine catabolism in the cyanobacterium Synechocystis sp. strain PCC6803 involves the urea cycle and arginase pathway. J. Bacteriol. 2000, 182, 1008–1015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Schreiber, S.; Konradt, M.; Groll, C.; Scheid, P.; Hanauer, G.; Werling, H.O.; Josenhans, C.; Suerbaum, S. The spatial orientation of Helicobacter pylori in the gastric mucus. Proc. Natl. Acad. Sci. USA 2004, 101, 5024–5029. [Google Scholar] [CrossRef] [Green Version]
  7. Eaton, K.A.; Morgan, D.R.; Krakowka, S. Campylobacter pylori virulence factors in gnotobiotic piglets. Infect. Immun. 1989, 57, 1119–1125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Tomb, J.F.; White, O.; Kerlavage, A.R.; Clayton, R.A.; Sutton, G.G.; Fleischmann, R.D.; Ketchum, K.A.; Klenk, H.P.; Gill, S.; Dougherty, B.A.; et al. The complete genome sequence of the gastric pathogen Helicobacter pylori. Nature 1997, 388, 539–547, Erratum in Nature 1997, 389, 412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Labigne, A.; De Reuse, H. Determinants of Helicobacter pylori pathogenicity. Infect. Agents Dis. 1996, 191–202. [Google Scholar] [CrossRef] [Green Version]
  10. Hatakeyama, M.; Higashi, H. Helicobacter pylori CagA: A new paradigm for bacterial carcinogenesis. Cancer Sci. 2005, 96, 835–843. [Google Scholar] [CrossRef]
  11. Roszczenko-Jasińska, P.; Wojtyś, M.I.; Jagusztyn-Krynicka, E.K. Helicobacter pylori treatment in the post-antibiotics era-searching for new drug targets. Appl. Microbiol. Biotechnol. 2020, 104, 9891–9905. [Google Scholar] [CrossRef]
  12. Tshibangu-Kabamba, E.; Yamaoka, Y. Helicobacter pylori infection and antibiotic resistance from biology to clinical implications. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 613–629. [Google Scholar] [CrossRef]
  13. WHO. Global Priority List of Antibiotic-Resistant Bacteria to Guide Research, Discovery, and Development of New Antibiotics. 2017. Available online: https://www.who.int/medicines/publications/WHO-PPL-Short_Summary_25Feb-ET_NM_WHO.pdf (accessed on 1 August 2021).
  14. Gerrits, M.; H M van Vliet, A.; Kuipers, E.; Kusters, J.G. Helicobacter pylori and antimicrobial resistance: Molecular mechanisms and clinical implications. Lancet Infect. Dis. 2006, 6, 699–709. [Google Scholar] [CrossRef]
  15. Thung, I.; Aramin, H.; Vavinskaya, V.; Gupta, S.; Park, J.Y.; Crowe, S.E.; Valasek, M.A. Review article: The global emergence of Helicobacter pylori antibiotic resistance. Aliment. Pharmacol. Ther. 2016, 43, 514–533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Nyssen, O.P.; Bordin, D.; Tepes, B.; Pérez-Aisa, Á.; Vaira, D.; Caldas, M.; Bujanda, L.; Castro-Fernandez, M.; Lerang, F.; Leja, M.; et al. European Registry on Helicobacter pylori management (Hp-EuReg): Patterns and trends in first-line empirical eradication prescription and outcomes of 5 years and 21 533 patients. Gut 2021, 70, 40–54. [Google Scholar] [CrossRef]
  17. Zamani, M.; Ebrahimtabar, F.; Zamani, V.; Miller, W.H.; Alizadeh-Navaei, R.; Shokri-Shirvani, J.; Derakhshan, M.H. Systematic review with meta-analysis: The worldwide prevalence of Helicobacter pylori infection. Aliment. Pharmacol. Ther. 2018, 47, 868–876. [Google Scholar] [CrossRef] [Green Version]
  18. 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] [Green Version]
  19. Sjomina, O.; Pavlova, J.; Niv, Y.; Leja, M. Epidemiology of Helicobacter pylori infection. Helicobacter 2018, 23, e12514. [Google Scholar] [CrossRef] [Green Version]
  20. Rowland, M.; Daly, L.; Vaughan, M.; Higgins, A.; Bourke, B.; Drumm, B. Age-specific incidence of Helicobacter pylori. Gastroenterology 2006, 130, 65–72; quiz 211. [Google Scholar] [CrossRef] [PubMed]
  21. Aguilera Matos, I.; Diaz Oliva, S.E.; Escobedo, A.A.; Villa Jiménez, O.M.; Velazco Villaurrutia, Y.D.C. Helicobacter pylori infection in children. BMJ Paediatr. Open 2020, 4, e000679. [Google Scholar] [CrossRef] [PubMed]
  22. Hoda, M.M.; Nancy, D.L.; David, Y.G.; Ramchatesingh, J.E. Helicobacter pylori Infection in Preschool and School-Aged Minority Children: Effect of Socioeconomic Indicators and Breast-Feeding Practices. Clin. Infect. Dis. 2001, 32, 1387–1392. [Google Scholar] [CrossRef] [Green Version]
  23. Thomas, J.E.; Austin, S.; Dale, A.; McClean, P.; Harding, M.; Coward, W.A.; Weaver, L.T. Protection by human milk IgA against Helicobacter pylori infection in infancy. Lancet 1993, 342, 121. [Google Scholar] [CrossRef]
  24. Sasaki, K.; Tajiri, Y.; Sata, M.; Fujii, Y.; Matsubara, F.; Zhao, M.G.; Shimizu, S.; Toyonaga, A.; Tanikawa, K. Helicobacter pylori in the natural environment. Scand. J. Infect. Dis. 1999, 31, 275–280. [Google Scholar]
  25. Velazquez, M.; Feirtag, J.M. Helicobacter pylori: Characteristics, pathogenicity, detection methods and mode of transmission implicating foods and water. Int. J. Food Microbiol. 1999, 53, 95–104. [Google Scholar] [CrossRef]
  26. Ansorg, R.; Vonheinegg, E.H.; Vonrecklinghausen, G. Cat owner’s risk of acquiring a Helicobacter pylori infection. Zentralbl. Bakteriol. Int. J. Med. Microbiol. Vir. Parasitol. Infect. Dis. 1995, 283, 122–126. [Google Scholar]
  27. Fox, J.G. Non-human reservoirs of Helicobacter pylori. Aliment. Pharmacol. Ther. 1995, 9, 93–103. [Google Scholar] [PubMed]
  28. Dore, M.P.; Bilotta, M.; Vaira, D.; Mancal, A.; Massarelli, G.; Leandro, G.; Atzei, A.; Pisanu, G.; Graham, D.Y.; Realdi, G. High prevalence of Helicobacter pylori infection in shepherds. Dig. Dis. Sci. 1999, 44, 1161–1164. [Google Scholar] [CrossRef] [PubMed]
  29. Goodman, K.J.; Correa, P. Transmission of Helicobacter pylori among siblings. Lancet 2000, 355, 358–362. [Google Scholar] [CrossRef]
  30. O’Connor, A.; O′Morain, C. Helicobacter pylori infection in Europe: Current perspectives. Expert Rev. Gastroenterol. Hepatol. 2013, 7, 541–548. [Google Scholar] [CrossRef]
  31. Kouitcheu Mabeku, L.B.; Noundjeu Ngamga, M.L.; Leundji, H. Potential risk factors and prevalence of Helicobacter pylori infection among adult patients with dyspepsia symptoms in Cameroon. BMC Infect. Dis. 2018, 18, 278. [Google Scholar] [CrossRef]
  32. Ogihara, A.; Kikuchi, S.; Hasegawa, A.; Kurosawa, M.; Miki, K.; Kaneko, E.; Mizukoshi, H. Relationship between Helicobacter pylori infection, smoking and drinking habits. J. Gastroenterol. Hepatol. 2000, 15, 271–276. [Google Scholar]
  33. Tonkic, A.; Tonkic, M.; Lehours, P.; Mégraud, F. Epidemiology and diagnosis of Helicobacter pylori infection. Helicobacter 2012, 17, 1–8. [Google Scholar] [CrossRef] [PubMed]
  34. Haley, K.P.; Gaddy, J.A. Nutrition and Helicobacter pylori: Host Diet and Nutritional Immunity Influence Bacterial Virulence and Disease Outcome. Gastroenterol. Res. Pract. 2016, 2016, 3019362. [Google Scholar] [CrossRef] [Green Version]
  35. Setiawan, V.W.; Zhang, Z.-F.; Yu, G.-P.; Lu, Q.Y.; Li, Y.L.; Lu, M.L.; Wang, M.R.; Guo, C.H.; Yu, S.Z.; Kurtz, R.C.; et al. Protective effect of green tea on the risks of chronic gastritis and stomach cancer. Int. J. Cancer 2001, 92, 600–604. [Google Scholar] [CrossRef] [PubMed]
  36. Lunet, N.; Lacerda-Vieira, A.; Barros, H. Fruit and vegetables consumption and gastric cancer: A systematic review and meta-analysis of cohort studies. Nutr. Cancer 2005, 53, 1–10. [Google Scholar] [CrossRef]
  37. Jarosz, M.; Dzieniszewski, J.; Dabrowska-Ufniarz, E.; Wartanowicz, M.; Ziemlanski, S.; Reed, P.I. Effects of high dose vitamin C treatment on Helicobacter pylori infection and total vitamin C concentration in gastric juice. Eur. J. Cancer Prev. 1998, 7, 449–454. [Google Scholar] [CrossRef]
  38. Venneman, K.; Huybrechts, I.; Gunter, M.J.; Vandendaele, L.; Herrero, R.; Van Herck, K. The epidemiology of Helicobacter pylori infection in Europe and the impact of lifestyle on its natural evolution toward stomach cancer after infection: A systematic review. Helicobacter 2018, 23, e12483. [Google Scholar] [CrossRef]
  39. Noto, J.M.; Gaddy, J.A.; Lee, J.Y. Iron deficiency accelerates Helicobacter pylori-induced carcinogenesis in rodents and humans. J. Clin. Investig. 2013, 123, 479–492. [Google Scholar] [CrossRef] [Green Version]
  40. Ghoshal, U.C.; Tiwari, S.; Dhingra, S. Frequency of Helicobacter pylori and CagA antibody in patients with gastric neoplasms and controls: The Indian enigma. Dig. Dis. Sci. 2008, 53, 1215–1222. [Google Scholar] [CrossRef] [PubMed]
  41. Khoder, G.; Muhammad, J.S.; Mahmoud, I.; Soliman, S.S.M.; Burucoa, C. Prevalence of Helicobacter pylori and Its Associated Factors among Healthy Asymptomatic Residents in the United Arab Emirates. Pathogens 2019, 8, 44. [Google Scholar] [CrossRef] [Green Version]
  42. World Cancer Research Fund; American Institute for Cancer Research (WCRF/AICR). Continuous Update Project Report: Diet, Nutrition, Physical Activity and Stomach Cancer. World Cancer Research Fund International: London, UK, 2016. Revised 2018. Available online: https://www.wcrf.org/wp-content/uploads/2021/02/stomach-cancer-report.pdf (accessed on 1 August 2021).
  43. Ibrahim, A.; Morais, S.; Ferro, A.; Lunet, N.; Peleteiro, B. Sex-differences in the prevalence of Helicobacter pylori infection in pediatric and adult populations: Systematic review and meta-analysis of 244 studies. Dig. Liver Dis. 2017, 49, 742–749. [Google Scholar] [CrossRef]
  44. Kim, H.J.; Choi, B.Y.; Byun, T.J.; Eun, C.S.; Song, K.S.; Kim, Y.S.; Han, D.S. The prevalence of atrophic gastritis and intestinal metaplasia according to gender, age and Helicobacter pylori infection in a rural population. J. Prev. Med. Public Health 2008, 41, 373–379. [Google Scholar] [CrossRef]
  45. Prashanth, R.; Barsouk, A. Epidemiology of gastric cancer: Global trends, risk factors and prevention. Prz. Gastroenterol. 2019, 14, 26–38. [Google Scholar]
  46. Muzaheed. Helicobacter pylori Oncogenicity: Mechanism, Prevention, and Risk Factors. Sci. World J. 2020, 10. [Google Scholar] [CrossRef]
  47. Saka, K.Y.; Nimura, S. Endoscopic and histological features of gastric cancers after successful Helicobacter pylori eradication therapy. Gastric Cancer 2016, 19, 524–530. [Google Scholar] [CrossRef]
  48. Setia, N.; Clark, J.W.; Duda, D.G.; Hong, T.S.; Kwak, E.L.; Mullen, J.T.; Lauwers, G.Y. Familial gastric cancers. Oncologist 2015, 20, 1365–1377. [Google Scholar] [CrossRef] [Green Version]
  49. Choi, Y.J.; Kim, N. Gastric cancer and family history. Korean J. Intern. Med. 2016, 31, 1042–1053. [Google Scholar] [CrossRef]
  50. Sasazuki, S.; Sasaki, S.; Tsugane, S. Cigarette smoking, alcohol consumption and subsequent gastric cancer risk by subsite and histologic type. Inter. J. Cancer 2002, 101, 560–566. [Google Scholar] [CrossRef]
  51. Wroblewski, L.E.; Peek, R.M.; Wilson, K.T. Helicobacter pylori and gastric cancer: Factors that modulate disease risk. Clin. Microbiol. Rev. 2010, 3, 713–739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Watari, J.; Chen, N.; Amenta, P.S.; Fukui, H.; Oshima, T.; Tomita, T.; Miwa, H.; Lim, K.J.; Das, K.M. Helicobacter pylori associated chronic gastritis, clinical syndromes, precancerous lesions, and pathogenesis of gastric cancer development. World J. Gastroenterol. 2014, 20, 5461–5473. [Google Scholar] [CrossRef] [PubMed]
  53. Stolte, M.; Bayerdörffer, E.; Morgner, A.; Alpen, B.; Wündisch, T.; Thiede, C.; Neubauer, A. Helicobacter and gastric MALT lymphoma. Gut 2002, 50, 3. [Google Scholar] [CrossRef] [PubMed]
  54. Matsumoto, H.; Shiotani, A.; Graham, D.Y. Current and Future Treatment of Helicobacter pylori Infections. Adv. Exp. Med. Biol. 2019, 1149, 211–225. [Google Scholar]
  55. Yang, J.C.; Lin, C.J. CYP2C19 genotypes in the pharmacokinetics/pharmacodynamics of proton pump inhibitor-based therapy of Helicobacter pylori infection. Expert Opin. Drug Metab. Toxicol. 2010, 6, 29–41. [Google Scholar] [CrossRef]
  56. Han, H.S.; Lee, S.Y. Correlations of the Gastric and Duodenal Microbiota with Histological, Endoscopic, and Symptomatic Gastritis. J. Clin. Med. 2019, 8, 312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Nezami, B.G.; Jani, M.; Alouani, D.; Rhoads, D.D.; Sadri, N. Helicobacter pylori Mutations Detected by Next-Generation Sequencing in Formalin-Fixed, Paraffin-Embedded Gastric Biopsy Specimens Are Associated with Treatment Failure. J. Clin. Microbiol. 2019, 57, e01834-18. [Google Scholar] [CrossRef] [Green Version]
  58. Ferreira, R.M.; Pereira-Marques, J.; Pinto-Ribeiro, I.; Costa, J.L.; Carneiro, F.; Machado, J.C.; Figueiredo, C. Gastric microbial community profiling reveals a dysbiotic cancer-associated microbiota. Gut 2018, 67, 226–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Malfertheiner, P.; Megraud, F.; O’Morain, C.A.; Gisbert, J.P.; Kuipers, E.J.; Axon, A.T.; Bazzoli, F.; Gasbarrini, A.; Atherton, J.; Graham, D.Y.; et al. European Helicobacter and Microbiota Study Group and Consensus panel. Management of Helicobacter pylori infection-the Maastricht V/Florence Consensus Report. Gut 2017, 66, 6–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Chey, W.D.; Wong, B.C. American College of Gastroenterology guideline on the management of Helicobacter pylori infection. Am. J. Gastroenterol. 2007, 102, 1808–1825. [Google Scholar] [CrossRef]
  61. Miftahussurur, M.; Cruz, M.; Subsomwong, P.; Jiménez Abreu, J.A.; Hosking, C.; Nagashima, H.; Akada, J.; Yamaoka, Y. Clarithromycin-Based Triple Therapy is Still Useful as an Initial Treatment for Helicobacter pylori Infection in the Dominican Republic. Am. J. Trop. Med. Hyg. 2017, 96, 1050–1059. [Google Scholar] [CrossRef] [Green Version]
  62. Gebeyehu, E.; Nigatu, D.; Engidawork, E. Helicobacter pylori eradication rate of standard triple therapy and factors affecting eradication rate at Bahir Dar city administration, Northwest Ethiopia: A prospective follow up study. PLoS ONE 2019, 14, e0217645. [Google Scholar] [CrossRef] [Green Version]
  63. Graham, D.Y.; Fischbach, L. Helicobacter pylori treatment in the era of increasing antibiotic resistance. Gut 2010, 59, 1143–1153. [Google Scholar] [CrossRef]
  64. Chung, J.W.; Han, J.P.; Kim, K.O.; Kim, S.Y.; Hong, S.J.; Kim, T.H.; Kim, C.W.; Kim, J.S.; Kim, B.W.; Bang, B.W.; et al. Ten-day empirical sequential or concomitant therapy is more effective than triple therapy for Helicobacter pylori eradication: A multicenter, prospective study. Dig. Liver. Dis. 2016, 48, 888–892. [Google Scholar] [CrossRef]
  65. Park, S.M.; Kim, J.S.; Kim, B.W.; Ji, J.S.; Choi, H. Randomized clinical trial comparing 10- or 14-day sequential therapy and 10- or 14-day concomitant therapy for the first line empirical treatment of Helicobacter pylori infection. J. Gastroenterol. Hepatol. 2017, 32, 589–594. [Google Scholar] [CrossRef]
  66. Ang, T.L.; Fock, K.M.; Song, M.; Ang, D.; Kwek, A.B.; Ong, J.; Tan, J.; Teo, E.K.; Dhamodaran, S. Ten-day triple therapy versus sequential therapy versus concomitant therapy as first-line treatment for Helicobacter pylori infection. J. Gastroenterol. Hepatol. 2015, 30, 1134–1139. [Google Scholar] [CrossRef]
  67. McNicholl, A.G.; Marin, A.C.; Molina-Infante, J.; Castro, M.; Barrio, J.; Ducons, J.; Calvet, X.; de la Coba, C.; Montoro, M.; Bory, F.; et al. Randomised clinical trial comparing sequential and concomitant therapies for Helicobacter pylori eradication in routine clinical practice. Gut 2014, 63, 244–249. [Google Scholar] [CrossRef]
  68. Gatta, L.; Vakil, N.; Vaira, D.; Scarpignato, C. Global eradication rates for Helicobacter pylori infection: Systematic review and meta-analysis of sequential therapy. BMJ 2013, 347, f4587. [Google Scholar] [CrossRef] [Green Version]
  69. He, L.; Deng, T.; Luo, H. Meta-analysis of sequential, concomitant and hybrid therapy for Helicobacter pylori eradication. Intern. Med. 2015, 54, 703–710. [Google Scholar] [CrossRef] [Green Version]
  70. Malfertheiner, P.; Selgrad, M. Helicobacter pylori infection and current clinical areas of contention. Curr. Opin. Gastroenterol. 2010, 26, 618–623. [Google Scholar] [CrossRef] [PubMed]
  71. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Hu, Y.; Zhu, Y.; Lu, N.H. Novel and Effective Therapeutic Regimens for Helicobacter pylori in an Era of Increasing Antibiotic Resistance. Front. Cell Infect. Microbiol. 2017, 5, 168. [Google Scholar] [CrossRef]
  73. Fallone, C.A.; Moss, S.F.; Malfertheiner, P. Reconciliation of Recent Helicobacter pylori Treatment Guidelines in a Time of Increasing Resistance to Antibiotics. Gastroenterology 2019, 157, 44–53. [Google Scholar] [CrossRef] [PubMed]
  74. Hsu, P.I.; Tsai, F.W.; Kao, S.S.; Hsu, W.H.; Cheng, J.S.; Peng, N.J.; Tsai, K.W.; Hu, H.M.; Wang, Y.K.; Chuah, S.K.; et al. Ten-day quadruple therapy comprising proton pump inhibitor, bismuth, tetracycline, and levofloxacin is more effective than standard levofloxacin triple therapy in the second-line treatment of Helicobacter pylori infection: A randomized controlled trial. Am. J. Gastroenterol. 2017, 112, 1374–1381. [Google Scholar] [CrossRef] [PubMed]
  75. Macias-Garcia, F.; Baston-Rey, I.; de la Iglesia-Garcia, D.; Calvino-Suarez, C.; Nieto-Garcia, L.; Dominguez-Munoz, J.E. Bismuth-containing quadruple therapy versus concomitant quadruple therapy as first-line treatment for Helicobacter pylori infection in an area of high resistance to clarithromycin: A prospective, cross-sectional, comparative, open trial. Helicobacter 2019, 24, e12546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Graham, D.Y.; Lee, S.Y. How to Effectively Use Bismuth Quadruple Therapy: The Good, the Bad, and the Ugly. Gastroenterol. Clin. N. Am. 2015, 44, 537–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Borsch, G.M.; Graham, D.Y. Helicobacter pylori. In Pharmacology of Peptic Ulcer Disease; Collen, M.J., Benjamin, S.B., Eds.; Handbook of Experimental Pharmacology; Springer: Berlin/Heidelberg, Germany, 1991; pp. 107–148. [Google Scholar]
  78. Xie, Y.; Pan, X.; Li, Y.; Wang, H.; Du, Y.; Xu, J.; Wang, J.; Zeng, Z.; Chen, Y.; Zhang, G. New single capsule of bismuth, metronidazole and tetracycline given with omeprazole versus quadruple therapy consisting of bismuth, omeprazole, amoxicillin and clarithromycin for eradication of Helicobacter pylori in duodenal ulcer patients: A Chinese prospective, randomized, multicentre trial. J. Antimicrob. Chemother. 2018, 73, 1681–1687. [Google Scholar] [PubMed]
  79. Fiorini, G.; Zullo, A.; Saracino, I.M.; Gatta, L.; Pavoni, M.; Vaira, D. Pylera and sequential therapy for first-line Helicobacter pylori eradication: A culture-based study in real clinical practice. Eur. J. Gastroenterol. Hepatol. 2018, 30, 621–625. [Google Scholar] [CrossRef] [PubMed]
  80. Andreev, D. Helicobacter pylori Eradication Therapy: Current Regimens. Adv. Res. Gastroenterol. Hepatol. 2017, 7, 555710. [Google Scholar] [CrossRef]
  81. Gisbert, J.P.; Calvet, X. Update on non-bismuth quadruple (concomitant) therapy for eradication of Helicobacter pylori. Clin. Exp. Gastroenterol. 2012, 5, 23–34. [Google Scholar] [CrossRef] [Green Version]
  82. Gisbert, J.P.; Calvet, X. Review article: Rifabutin in the treatment of refractory Helicobacter pylori infection. Aliment. Pharmacol. Ther. 2012, 35, 209–221. [Google Scholar] [CrossRef]
  83. Bock, H.M.D.; Koop, H.M.D.; Lehn, N.M.D.; Heep, M.M.D. Rifabutin-based Triple Therapy After Failure of Helicobacter pylori Eradication Treatment: Preliminary Experience. J. Clin. Gastroenterol. 2000, 31, 222–225. [Google Scholar] [CrossRef]
  84. Ribaldone, D.G.; Fagoonee, S.; Astegiano, M. Rifabutin-Based Rescue Therapy for Helicobacter pylori Eradication: A Long-Term Prospective Study in a Large Cohort of Difficult-to-Treat Patients. J. Clin. Med. 2019, 8, 199. [Google Scholar] [CrossRef] [Green Version]
  85. Zhang, M.; Chen, C.Y.; Wang, X.T.; Lyu, B. Levofloxacin-based triple therapy versus bismuth-based quadruple therapy in the treatment of Helicobacter pylori as the rescue therapy: A meta analysis. Zhonghua Nei Ke Za Zhi 2017, 56, 368–374. [Google Scholar]
  86. Glocker, E.; Stueger, H.P.; Kist, M. Quinolone resistance in Helicobacter pylori isolates in Germany. Antimicrob. Agents Chemother. 2007, 51, 346–349. [Google Scholar] [CrossRef] [Green Version]
  87. Kang, J.M.; Kim, N.; Lee, D.H.; Park, Y.S.; Kim, Y.R.; Kim, J.S. Second-line treatment for Helicobacter pylori infection: 10-day moxifloxacin-based triple therapy versus 2-week quadruple therapy. Helicobacter 2007, 12, 623–628. [Google Scholar] [CrossRef]
  88. Nista, E.C.; Candelli, M.; Zocco, M.A.; Cazzato, I.A.; Cremonini, F.; Ojetti, V. Moxifloxacin-based strategies for first-line treatment of Helicobacter pylori infection. Aliment. Pharmacol. 2005, 21, 1241–1247. [Google Scholar] [CrossRef] [PubMed]
  89. Berning, M.; Krasz, S.; Miehlke, S. Should quinolones come first in Helicobacter pylori therapy? Therap. Adv. Gastroenterol. 2011, 4, 103–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Gupta, V.K.; Dhar, A.; Srinivasan, S.; Rattan, A.; Sharma, M.P. Eradication of H. pylori in a developing country: Comparison of lansoprazole versus omeprazole with norfloxacin, in a dual-therapy study. Am. J. Gastroenterol. 1997, 92, 1140–1142. [Google Scholar] [PubMed]
  91. Zaidi, S.F.; Muhammad, J.S.; Usmanghani, K.; Sugiyama, T. Pharmacological ins and outs of medicinal plants against Helicobacter pylori: A review. Pak. J. Pharm. Sci. 2015, 2, 28. [Google Scholar]
  92. Hunt, R.H.; Scarpignato, C. Potassium-Competitive Acid Blockers (P-CABs): Are They Finally Ready for Prime Time in Acid-Related Disease? Clin. Transl. Gastroenterol. 2015, 6, e119. [Google Scholar] [CrossRef]
  93. Tanabe, H.; Ando, K.; Sato, K.; Ito, T.; Goto, M.; Sato, T.; Fujinaga, A.; Kawamoto, T.; Utsumi, T.; Yanagawa, N. Efficacy of Vonoprazan-Based Triple Therapy for Helicobacter pylori Eradication: A Multicenter Study and a Review of the Literature. Dig. Dis. Sci. 2017, 62, 3069–3076. [Google Scholar] [CrossRef]
  94. Njume, C.; Afolayan, A.J.; Ndip, R.N. An overview of antimicrobial resistance and the future of medicinal plants in the treatment of Helicobacter pylori infections. Afr. J. Pharm. Pharmacol. 2009, 3, 685–699. [Google Scholar]
  95. Safavi, M.; Shams-Ardakani, M.; Foroumadi, A. Medicinal plants in the treatment of Helicobacter pylori infections. Pharm. Biol. 2015, 53, 939–960. [Google Scholar] [CrossRef]
  96. O’Gara, E.A.; Hill, D.J.; Maslin, D.J. Activities of garlic oil, garlic powder, and their diallyl constituents against Helicobacter pylori. Appl. Environ. Microbiol. 2000, 66, 2269–2273. [Google Scholar] [CrossRef] [Green Version]
  97. O’Gara, E.A.; Maslin, D.J.; Nevill, A.M.; Hill, D.J. The effect of simulated gastric environments on the anti-Helicobacter activity of garlic oil. J. Appl. Microbiol. 2007, 104, 1324–1331. [Google Scholar] [CrossRef] [PubMed]
  98. Xiao, S.; Zhang, X.-M. Allicin as add-on therapy for Helicobacter pylori infection: A systematic review and meta-analysis. World J. Gastroenterol. 2019, 25, 6025–6040. [Google Scholar] [CrossRef]
  99. Okeleye, B.I.; Bessong, P.O.; Ndip, R.N. Preliminary phytochemical screening and in vitro anti-Helicobacter pylori activity of extracts of the stem bark of Bridelia micrantha (Hochst., Baill., Euphorbiaceae). Molecules 2011, 16, 6193–6205. [Google Scholar] [CrossRef] [PubMed]
  100. Adefuye, A.O.; Ndip, R.N. Phytochemical analysis and antibacterial evaluation of the ethyl acetate extract of the stem bark of Bridelia micrantha. Pharmacogn. Mag. 2013, 9, 45–50. [Google Scholar] [CrossRef] [Green Version]
  101. Baker, D.A. Plants against Helicobacter pylori to combat resistance: An ethnopharmacological review. Biotechnol. Rep. 2020, 26, e00470. [Google Scholar] [CrossRef] [PubMed]
  102. Ruggiero, P.; Rossi, G.; Tombola, F. Red wine and green tea reduce H pylori- or VacA-induced gastritis in a mouse model. World J. Gastroenterol. 2007, 13, 349–354. [Google Scholar] [CrossRef] [Green Version]
  103. Shoae Hassani, A.; Ordouzadeh, N.; Ghaemi, A.; Amirmozafari, N.; Hamdi, K.; Nazari, R. In vitro inhibition of Helicobacter pylori urease with non and semi fermented Camellia sinensis. Indian J. Med. Microbiol. 2009, 27, 30–34. [Google Scholar] [CrossRef]
  104. Boyanova, L.; Ilieva, J.; Gergova, G.; Vladimirov, B.; Nikolov, R.; Mitova, I. Honey and green/black tea consumption may reduce the risk of Helicobacter pylori infection. Diagn. Microbiol. Infect. Dis. 2015, 82, 85–86. [Google Scholar] [CrossRef] [PubMed]
  105. Bergonzelli, G.E.; Donnicola, D.; Porta, N.; Corthésy-Theulaz, I.E. Essential oils as components of a diet-based approach to management of Helicobacter infection. Antimicrob. Agents Chemother. 2003, 47, 3240–3246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Asdaq, S.M.B.; Swathi, E.; Dhamanigi, S.S. Role of Daucus carota in Enhancing Antiulcer Profile of Pantoprazole in Experimental Animals. Molecules 2020, 25, 5287. [Google Scholar] [CrossRef] [PubMed]
  107. Shaik, M.A.; Aleem, A.K.; Irshad, A.; Musaddiq, M.; Khaja, S.A.; Polasa, H.; L Venkateswar, R.; Chittoor, M.H.; Leonardo, A.S.; Niyaz, A. Antimicrobial activities of Eugenol and Cinnamaldehyde against the human gastric pathogen Helicobacter pylori. Ann. Clin. Microbiol. Antimicrob. 2005, 4, 20. [Google Scholar] [CrossRef] [Green Version]
  108. Wang, Y.C. Medicinal plant activity on Helicobacter pylori related diseases. World J. Gastroenterol. 2014, 20, 10368–10382. [Google Scholar] [CrossRef] [PubMed]
  109. Shahani, S.; Monsef-Esfahani, H.R.; Saeidnia, S.; Sanie, P.; Siavoshi, F.; Foroumadi, A.; Samadi, N.; Gohari, A.R. Anti-Helicobacter pylori activity of the methanolic extract of Geum iranicum and its main compounds. Z. Naturforsch. C 2012, 67, 172–180. [Google Scholar] [CrossRef]
  110. De Mello Moraes, T.; Rodrigues, C.M.; Kushima, H.; Bauab, T.M.; Villegas, W.; Pellizzon, C.H.; Brito, A.R.; Hiruma-Lima, C.A. Hancornia speciosa: Indications of gastroprotective, healing and anti-Helicobacter pylori actions. J. Ethnopharmacol. 2008, 120, 161–168. [Google Scholar] [CrossRef]
  111. Xavier Bastos, K.; Noleto Dias, C.; Mangueira Nascimento, Y.; Sobral da Silva, M.; Zucolotto Langassner, S.M.; Wessjohann, L.A.; Fechine Tavares, J. Identification of Phenolic Compounds from Hancornia speciosa (Apocynaceae) Leaves by UHPLC Orbitrap-HRMS. Molecules 2017, 22, 143. [Google Scholar] [CrossRef] [Green Version]
  112. Deriu, A.; Branca, G.; Molicotti, P.; Pintore, G.; Chessa, M.; Tirillini, B.; Paglietti, B.; Mura, A.; Sechi, L.A.; Fadda, G.; et al. In vitro activity of essential oil of Myrtus communis L. against Helicobacter pylori. Inter. J. Antimicrob. Agents 2007, 30, 562–563. [Google Scholar] [CrossRef]
  113. Sisay, M.; Gashaw, T. Ethnobotanical, Ethnopharmacological, and Phytochemical Studies of Myrtus communis Linn: A Popular Herb in Unani System of Medicine. J. Evid.-Based Compl. Altern. Med. 2017, 22, 1035–1043. [Google Scholar] [CrossRef] [Green Version]
  114. Pastene, E.; Speisky, H.; García, A.; Moreno, J.; Troncoso, M.; Figueroa, G. In vitro and in vivo effects of apple peel polyphenols against Helicobacter pylori. J. Agricul. Food Chem. 2010, 58, 7172–7179. [Google Scholar] [CrossRef]
  115. Salehi, B.; Sharopov, F.; Martorell, M.; Rajkovic, J.; Ademiluyi, A.O.; Sharifi-Rad, M.; Fokou, P.V.T.; Martins, N.; Iriti, M.; Sharifi-Rad, J. Phytochemicals in Helicobacter pylori Infections: What Are We Doing Now? Int. J. Mol. Sci. 2018, 19, 2361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Sudjana, A.N.; D’Orazio, C.; Ryan, V.; Rasool, N.; Ng, J.; Islam, N.; Riley, T.V.; Hammer, K.A. Antimicrobial activity of commercial Olea europaea (olive) leaf extract. Int. J. Antimicrob. Agents 2009, 33, 461–463. [Google Scholar] [CrossRef] [PubMed]
  117. Lee, O.-H.; Lee, B.-Y. Antioxidant and antimicrobial activities of individual and combined phenolics in Olea europaea leaf extract. Bioresour. Technol. 2010, 101, 3751–3754. [Google Scholar] [CrossRef] [PubMed]
  118. Bisignano, C.; Filocamo, A.; La Camera, E.; Zummo, S.; Fera, M.T.; Mandalari, G. Antibacterial activities of almond skins on cagA-positive and-negative clinical isolates of Helicobacter pylori. BMC Microbiol. 2013, 13, 1–6. [Google Scholar] [CrossRef] [Green Version]
  119. Musarra-Pizzo, M.; Ginestra, G.; Smeriglio, A.; Pennisi, R.; Sciortino, M.T.; Mandalari, G. The Antimicrobial and Antiviral Activity of Polyphenols from Almond (Prunus dulcis L.) Skin. Nutrients 2019, 11, 2355. [Google Scholar] [CrossRef] [Green Version]
  120. Han, J.H.; Khin, P.P.; Sohn, U.D. Effect of Rumex aquaticus herba extract against Helicobacter pylori-induced inflammation in gastric epithelial cells. J. Medic. Food 2016, 19, 31–37. [Google Scholar] [CrossRef]
  121. Kwak, H.; Park, S.; Nguyen, Y.T.T.; Kim, C.H.; Lee, J.M.; Suh, J.S.; Whang, W.K.; Sohn, U.D. Protective Effect of Extract from Rumex Aquaticus Herba on Ethanol-induced Gastric Damage in Rats. Pharmacology 2012, 90, 288–297. [Google Scholar] [CrossRef]
  122. Khanavi, M.; Safavi, M.; Siavoshi, F.; Fallah Tafti, A.; Hajimahmoodi, M.; Hadjiakhoondi, A.; Rezazadeh, S.; Foroumadi, A. Evaluation of anti-Helicobacter pylori activity of methanol extracts of some species of Stachys and Melia. J. Med. Plants 2008, 4, 74–80. [Google Scholar]
  123. Nostro, A.; Cellini, L.; Bartolomeo, S.D.; Cannatelli, M.A.; Campli, E.D.; Procopio, F.; Grande, R.; Marzio, L.; Alonzo, V. Effects of combining extracts (from propolis or Zingiber officinale) with clarithromycin on Helicobacter pylori. Phytother. Res. Int. J. Devoted Pharmacol. Toxicol. Eval. Nat. Prod. Deriv. 2006, 20, 187–190. [Google Scholar]
  124. Siddaraju, M.N.; Shylaja, M.D. Inhibition of gastric H+,K+-ATPase and Helicobacter pylori growth by phenolic antioxidants of Zingiber officinale. Mol. Nutr. Food Res. 2007, 51, 324–332. [Google Scholar] [CrossRef]
  125. Ebrahimzadeh Attari, V.; Somi, M.H.; Asghari Jafarabadi, M.; Ostadrahim, A.; Moaddab, S.Y.; Lotfi, N. The Gastro-protective Effect of Ginger (Zingiber officinale Roscoe) in Helicobacter pylori Positive Functional Dyspepsia. Adv. Pharm. Bull. 2019, 9, 321–324. [Google Scholar] [CrossRef]
  126. Sforcin, J.M.; Bankova, V. Propolis: Is there a potential for the development of new drugs? J. Ethnopharmacol. 2011, 133, 253–260. [Google Scholar] [CrossRef]
  127. Salatino, A.; Teixeira, E.W.; Negri, G.; Message, D. Origin and chemical variation of Brazilian propolis. Evid.-Based Complementary Altern. Med. 2005, 2, 33–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Shapla, U.M.; Raihan, J.I.A.; Alam, F.; Solayman, N.; Gan, S.H.; Hossen, S.; Khalil, I. Propolis: The future therapy against Helicobacter pylori-mediated gastrointestinal diseases. J. Appl. Biomed. 2018, 16, 81–99. [Google Scholar] [CrossRef] [Green Version]
  129. Baltas, N.; Karaoglu, S.A.; Tarakci, C.; Kolayli, S. Effect of propolis in gastric disorders: Inhibition studies on the growth of Helicobacter pylori and production of its urease. J. Enzym. Inhib. Med. Chem. 2016, 31, 46–50. [Google Scholar] [CrossRef] [Green Version]
  130. Boyanova, L.; Gergova, G.; Nikolov, R.; Derejian, S.; Lazarova, E.; Katsarov, N. Activity of Bulgarian propolis against 94 Helicobacter pylori strains in vitro by agar-well diffusion, agar dilution and disc diffusion methods. J. Med. Microbiol. 2005, 54, 481–483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Coelho, L.; Bastos, E.; Resende, C.; Paula e Silva, C.; Sanches, B.; de Castro, F.; Moretzsohn, L.D.; Dos Santos Vieira, W.L.; Trindade, O.R. Brazilian green propolis on Helicobacter pylori infection. A pilot clinical study. Helicobacter 2007, 12, 572–574. [Google Scholar] [CrossRef] [PubMed]
  132. Mazel, D.; Pochet, S.; Marliere, P. Genetic characterization of polypeptide deformylase, a distinctive enzyme of eubacterial translation. EMBO J. 1994, 13, 914–923. [Google Scholar] [CrossRef] [PubMed]
  133. Ben-Hamida, A.; Man, W.K.; McNeil, N.; Spencer, J. Histamine, xanthine oxidase generated oxygen-derived free radicals and Helicobacter pylori in gastroduodenal inflammation and ulceration. Inflamm. Res. 1998, 47, 193–199. [Google Scholar] [CrossRef]
  134. Galeotti, F.; Maccari, F.; Fachini, A.; Volpi, N. Chemical Composition and Antioxidant Activity of Propolis Prepared in Different Forms and in Different Solvents Useful for Finished Products. Foods 2018, 7, 41. [Google Scholar] [CrossRef] [Green Version]
  135. Radha, K.M.; Anoop, K.S.; Jaya, G.; Rikhab, C.S. Multiple biological activities of curcumin: A short review. Life Sci. 2006, 78, 2081–2087. [Google Scholar]
  136. Sarkar, A.; De, R.; Mukhopadhyay, A.K. Curcumin as a potential therapeutic candidate for Helicobacter pylori associated diseases. World J. Gastroenterol. 2016, 22, 2736. [Google Scholar] [CrossRef] [PubMed]
  137. Irving, G.R.; Karmokar, A.; Berry, D.P.; Brown, K.; Steward, W.P. Curcumin: The potential for efficacy in gastrointestinal diseases. Best Pract. Res. Clin. Gastroenterol. 2011, 25, 519–534. [Google Scholar] [CrossRef]
  138. Jurenka, J.S. Anti-inflammatory properties of curcumin, a major constituent of Curcuma longa: A review of preclinical and clinical research. Altern. Med. Rev. 2009, 14, 143–153. [Google Scholar]
  139. Kumar, D.; Kumar, M.; Saravanan, C.; Singh, S.K. Curcumin: A potential candidate for matrix metalloproteinase inhibitors. Expert Opin. Ther. Targets 2012, 16, 959–972. [Google Scholar] [CrossRef]
  140. Marathe, S.A.; Sen, M.; Dasgupta, I.; Chakravortty, D. Differential modulation of intracellular survival of cytosolic and vacuolar pathogens by curcumin. Antimicrob. Agents Chemother. 2012, 56, 5555–5567. [Google Scholar] [CrossRef] [Green Version]
  141. Vetvicka, V.; Vetvickova, J.; Fernandez-Botran, R. Effects of curcumin on Helicobacter pylori infection. Ann. Transl. Med. 2016, 4, 479, Erratum in Ann Transl. Med. 2017, 5, 153. [Google Scholar] [CrossRef] [Green Version]
  142. Kim, Y.R.; Yang, C.S. Protective roles of ginseng against bacterial infection. Microb. Cell 2018, 5, 472. [Google Scholar] [CrossRef] [PubMed]
  143. Park, S.; Yeo, M.; Jin, J.H.; Lee, K.M.; Jung, J.Y.; Choue, R.; Cho, S.W.; Hahm, K.B. Rescue of Helicobacter pylori–induced cytotoxicity by red ginseng. Dig. Dis. Sci. 2005, 50, 1218–1227. [Google Scholar] [CrossRef] [Green Version]
  144. Cho, W.C.; Chung, W.S.; Lee, S.K.; Leung, A.W.; Cheng, C.H.; Yue, K.K. Ginsenoside Re of Panax ginseng possesses significant antioxidant and antihyperlipidemic efficiency in streptozotocin-induced diabetic rats. Eur. J. Pharmacol. 2006, 550, 173–179. [Google Scholar] [CrossRef] [PubMed]
  145. Bae, M.; Jang, S.; Lim, J.W. Protective effect of Korean Red Ginseng extract against Helicobacter pylori-induced gastric inflammation in Mongolian gerbils. J. Ginseng Res. 2014, 38, 8–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Wee, J.J.; Mee, P.K.; Chung, A.S. Biological Activities of Ginseng and Its Application to Human Health. In Herbal Medicine: Biomolecular and Clinical Aspects, 2nd ed.; Benzie, I.F.F., Wachtel-Galor, S., Eds.; CRC Press/Taylor & Francis: Boca Raton, FL, USA, 2011. Available online: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/books/NBK92776/ (accessed on 1 August 2021).
  147. Hyunju, K.J.; Weon, L.; Hyeyoung, K. Inhibitory effect of Korean Red Ginseng extract on DNA damage response and apoptosis in Helicobacter pylori–infected gastric epithelial cells. J. Ginseng Research. 2020, 1, 79–85. [Google Scholar]
  148. Singh, V.K.; Singh, D.K. Pharmacological Effects of Garlic (Allium sativum L.). Annu. Rev. Biomed. Sci. 2008, 10, 6–26. [Google Scholar] [CrossRef]
  149. Ross, S.; Feldberg Chang, S.C.; Kotik, A.N.; Nadler, M.; Neuwirth, Z.; Sundstrom, D.C.; Thompson, N.H. In Vitro Mechanism of Inhibition of Bacterial Cell Growth by Allicin. Antimicrob. Agents Chemother. 1988, 12, 1763–1768. [Google Scholar]
  150. Ejaz, S.; Woong, L.C.; Ejaz, A. Extract of garlic (Allium sativum) in cancer chemoprevention. Experiment. Oncol. 2003, 25, 93–97. [Google Scholar]
  151. Islam, M.S.; Kusumoto, Y.; Al-Mamun, M.A. Cytotoxicity and Cancer (HeLa) Cell Killing Efficacy of Aqueous Garlic (Allium sativum) Extract. J. Sci. Res. 2011, 3, 375–382. [Google Scholar] [CrossRef] [Green Version]
  152. Capasso, A. Antioxidant action and therapeutic efficacy of Allium sativum L. Molecules 2013, 18, 690–700. [Google Scholar] [CrossRef]
  153. Zhang, Q.; Wu, W.; Zhang, J.; Xia, X. Eradication of Helicobacter pylori: The power of nanosized formulations. Nanomedicine 2020. [Google Scholar] [CrossRef]
  154. Fritea, L.; Banica, F.; Costea, T.O.; Moldovan, L.; Iovan, C.; Cavalu, S. A gold nanoparticles—Graphene based electrochemical sensor for sensitive determination of nitrazepam. J. Electroanalyt. Chem. 2018, 830–831, 63–71. [Google Scholar] [CrossRef]
  155. Vala, A.K.; Trivedi, H.; Gosai, H.; Panseriya, H.; Dave, B. Biosynthesized silver nanoparticles and their therapeutic applications. Compr. Analyt. Chem. 2021, 94, 547–584. [Google Scholar] [CrossRef]
  156. Roy, A. Plant Derived Silver Nanoparticles and their Therapeutic Applications. Curr. Pharm. Biotechnol. 2020. [Google Scholar] [CrossRef]
  157. Gurunathan, S.; Jeong, J.K.; Han, J.W.; Zhang, X.F.; Park, J.H.; Kim, J.H. Multidimensional effects of biologically synthesized silver nanoparticles in Helicobacter pylori, Helicobacter felis, and human lung (L132) and lung carcinoma A549 cells. Nanoscale Res. Lett. 2015, 10, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Mikhailova, E.O. Silver Nanoparticles: Mechanism of Action and Probable Bio-Application. J. Funct. Biomater. 2020, 11, 84. [Google Scholar] [CrossRef] [PubMed]
  159. Amin, M.; Anwar, F.; Janjua, M.R.; Iqbal, M.A.; Rashid, U. Green synthesis of silver nanoparticles through reduction with Solanum xanthocarpum L. berry extract: Characterization, antimicrobial and urease inhibitory activities against Helicobacter pylori. Int. J. Mol. Sci. 2012, 13, 9923–9941. [Google Scholar] [CrossRef]
  160. Safarov, T.; Kiran, B.; Bagirova, M.; Allahverdiyev, A.M.; Abamor, E.S. An overview of nanotechnology-based treatment approaches against Helicobacter pylori. Expert Rev. Anti-Infect. Ther. 2019. [Google Scholar] [CrossRef]
  161. Valizadeh, H.; Mohammadi, G.; Ehyaei, R.; Milani, M.; Azhdarzadeh, M.; Zakeri-Milani, P.; Lotfipour, F. Antibacterial activity of clarithromycin loaded PLGA nanoparticles. Die Pharm. Int. J. Pharm. Sci. 2012, 67, 63–68. [Google Scholar]
  162. Lotfipour, F.; Valizadeh, H.; Milani, M.; Bahrami, N.; Ghotaslou, R. Study of antimicrobial effects of clarithromycin loaded PLGA nanoparticles against clinical strains of Helicobacter pylori. Drug Res. 2016, 66, 41–45. [Google Scholar] [CrossRef]
  163. Ramteke, S.; Jain, N.K. Clarithromycin-and omeprazole-containing gliadin nanoparticles for the treatment of Helicobacter pylori. J. Drug Target. 2008, 16, 65–72. [Google Scholar] [CrossRef]
  164. Pan-In, P.; Banlunara, W.; Chaichanawongsaroj, N.; Wanichwecharungruang, S. Ethyl cellulose nanoparticles: Clarithomycin encapsulation and eradication of H. pylori. Carbohydr. Polym. 2014, 109, 22–27. [Google Scholar] [CrossRef] [PubMed]
  165. Cavalu, S.; Bisboaca, S.; Mates, I.M.; Pasca, P.M.; Laslo, V.; Costea, T.; Fritea, L.; Vicas, S. Novel Formulation Based on Chitosan-Arabic Gum Nanoparticles Entrapping Propolis Extract: Production, physico-chemical and structural characterization. Rev. Chim. 2018, 69, 3756–3760. [Google Scholar] [CrossRef]
  166. Gonçalves, I.C.; Henriques, P.C.; Seabra, C.L.; Martins, M.C. The potential utility of chitosan micro/nanoparticles in the treatment of gastric infection. Expert Rev. Anti-Infect. Ther. 2014, 12, 981–992. [Google Scholar] [CrossRef]
  167. Lin, Y.-H.; Chang, C.-H.; Wu, Y.-S.; Hsu, Y.-M.; Chiou, S.-F.; Chen, Y.-J. Development of pH-responsive chitosan/heparin nanoparticles for stomach-specific anti-Helicobacter pylori therapy. Biomaterials 2009, 30, 3332–3342. [Google Scholar] [CrossRef]
  168. Hamman, J.H. Chitosan based polyelectrolyte complexes as potential carrier materials in drug delivery systems. Mar. Drugs 2010, 8, 1305–1322. [Google Scholar] [CrossRef] [Green Version]
  169. Bardonnet, P.-L.; Faivre, V.; Boullanger, P.; Piffaretti, J.-C.; Falson, F. Pre-formulation of liposomes against Helicobacter pylori: Characterization and interaction with the bacteria. Eur. J. Pharm. Biopharm. 2008, 69, 908–922. [Google Scholar] [CrossRef] [PubMed]
  170. Jung, S.W.; Thamphiwatana, S.; Zhang, L.; Obonyo, M. Mechanism of antibacterial activity of liposomal linolenic acid against Helicobacter pylori. PLoS ONE 2015, 10, e0116519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Thamphiwatana, S.; Gao, W.; Obonyo, M.; Zhang, L. In vivo treatment of Helicobacter pylori infection with liposomal linolenic acid reduces colonization and ameliorates inflammation. Proc. Natl. Acad. Sci. USA 2014, 111, 17600–17605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Seabra, C.L.; Nunes, C.; Gomez-Lazaro, M.; Correia, M.; Machado, J.C.; Gonçalves, I.C.; Reis, C.A.; Reis, S.; Martins, M.C.L. Docosahexaenoic acid loaded lipid nanoparticles with bactericidal activity against Helicobacter pylori. Int. J. Pharm. 2017, 519, 128–137. [Google Scholar] [CrossRef]
  173. Thamphiwatana, S.; Gao, W.; Pornpattananangkul, D.; Zhang, Q.; Fu, V.; Li, J.; Obonyo, M.; Zhang, L. Phospholipase A2-responsive antibiotic delivery via nanoparticle-stabilized liposomes for the treatment of bacterial infection. J. Mater. Chem. B 2014, 2, 8201–8207. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Carcinogenic effect of H. pylori through different mechanisms.
Figure 1. Carcinogenic effect of H. pylori through different mechanisms.
Molecules 26 06078 g001
Figure 2. H. pylori infections and alternative treatment approaches.
Figure 2. H. pylori infections and alternative treatment approaches.
Molecules 26 06078 g002
Table 1. Plants and their formulations as active agents against H. pylori.
Table 1. Plants and their formulations as active agents against H. pylori.
Name/FamilyFormulationMain Outcomes In Vitro/In VivoReference
Allium sativum/
Amaryllidaceae
Garlic oil and
powder
In vitro (standard and clinical isolates)—significant loss of viability, inactivation irrespective of strain, depending on allicin concentration
In vivo (clinical trials)-improves eradication rates, healing rates of ulcers, remission of peptic ulcers, synergism with conventional therapy
[96,97,98]
Bridelia micrantha/
Euphorbiaceous
Stem bark extractIn vitro (standard strains and clinical isolates)—killing rate depending on concentration and time (agar-well diffusion and MIC method)
Antioxidant, anti-inflammatory properties
[99,100,101]
Camellia sinensis/
Theaceae
Young shoot
extract
In vivo (animal model)—prevents gastritis, limits the localization of bacteria and VacA to the surface of the gastric epithelium. In vivo (clinical trial)—suppression of gastritis and prevalence of H. pylori in a dose-dependent manner.
In vitro—inhibits the production and function of the urease enzyme.
[102,103,104]
Daucus carota/
Apiaceae
Essential oil of
carrot seed
In vitro and in vivo (animal model)—co-administration with pantoprazole exhibits strong anti-H. pylori activity[105,106]
Eugenia caryophillus/
Myrtaceae
Extracts of
flowers
In vitro (standard strains and clinical isolates)—decrease viability of H. pylori, irrespective of the strain, at acidic pH, effective in gastritis[107,108]
Geum iranicum/
Rosaceae
Aqueous extract
of roots
In vitro- effective against clinical isolates of H. pylori resistant to metronidazole[109]
Hancornia speciosa/
Apocyanaceae
Bark’s hydroalcoholic
extract
In vivo (rodent model)- anti-H. pylori effect by increasing of gastric mucus formation and antioxidant properties of polymeric pro-anthocyanidins
Anti-inflammatory, antibacterial, and anticancer properties.
[110,111]
Myrtus communis/
Myrtaceae
Essential oil
extract
In vitro—effective against clinically isolated strains resistant to nitroimidazoles and clarithromycin.
Antimicrobial, antidiarrheal, anticancer, antioxidant, antiulcer, anti-inflammatory and antidiabetic activity
[112,113]
Malus domestica/
Rosaceae
Apple peel
extract
In vitro—prevents vacuolation in HeLa cells, antiadhesive effect against H. pylori.
In vivo (short-term infection model mice)—inhibitory effect on H. pylori attachment, anti-inflammatory effect on H. pylori-associated gastritis, reversible inhibitory effect
[114,115]
Olea europaea/
Oleaceae
Olive leaf
extract
In vitro (clinically isolated meticillin resistant strains)—regulates the composition of the gastric flora, selectively reduce levels of H. pylori
Antiulcer, antioxidant and antimicrobial effects
[116,117]
Prunus dulcis/
Rosaceae
Polyphenol
extract
In vitro (standard strains and clinical isolates)—effective against H. pylori with different virulence irrespective of the cagA and vacA status.[118,119]
Rumex aquaticus/
Polygonaceae
Plant extractIn vitro (human adenocarcinoma gastric cells)—cytotoxic effects against AGS cells, inhibit the production of proinflammatory cytokines, antioxidant[120,121]
Stachys setifera/
Lamiaceae
Aerial part
extract
In vitro (isolates)—strong inhibitory effect[101,122]
Zingiber officinale/
Zingiberaceae
Rhizome extractIn vitro—potent inhibitors of proton potassium ATPase activity and H. pylori growth.
In vivo (pilot study)—significant H. pylori eradication rate in dyspeptic patients, additive effect along with antibiotics
[123,124,125]
Propolis/
Apis melifera
Ethanolic extractsIn vitro—anti-H. pylori and anti-urease activities, 92.1% inhibition rate, synergism with clarithromycin or metronidazole[126,127,128,129,130,131,132,133,134]
Curcuma longa/
Zingiberaceae
Rhizome extract,
polyphenolicrich
extract of the root
In vitro (clinical isolates)—stabilizes the vacuolar membrane and prevents the escape of cytosolic pathogens, suppressor of the type 1 immune response.
In vivo (mouse model)—increase IL-4 serum levels and IgG, decrease IFN-γ levels
[135,136,137,138,139,140,141]
Ginseng/
Panax ginseng Meyer
Korean Red Ginseng
extract
In vitro (infected gastric epithelial cells)—reduces reactive oxygen species and prevents cell death
In vivo (animal model Mongolian gerbils)—anti-inflammatory effect on H. pylori-induced gastric inflammation
[142,143,144,145,146,147]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cardos, I.A.; Zaha, D.C.; Sindhu, R.K.; Cavalu, S. Revisiting Therapeutic Strategies for H. pylori Treatment in the Context of Antibiotic Resistance: Focus on Alternative and Complementary Therapies. Molecules 2021, 26, 6078. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26196078

AMA Style

Cardos IA, Zaha DC, Sindhu RK, Cavalu S. Revisiting Therapeutic Strategies for H. pylori Treatment in the Context of Antibiotic Resistance: Focus on Alternative and Complementary Therapies. Molecules. 2021; 26(19):6078. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26196078

Chicago/Turabian Style

Cardos, Ioana Alexandra, Dana Carmen Zaha, Rakesh K. Sindhu, and Simona Cavalu. 2021. "Revisiting Therapeutic Strategies for H. pylori Treatment in the Context of Antibiotic Resistance: Focus on Alternative and Complementary Therapies" Molecules 26, no. 19: 6078. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26196078

Article Metrics

Back to TopTop