Next Article in Journal
Influence of Bone Morphology on In Vivo Tibio-Femoral Kinematics in Healthy Knees during Gait Activities
Next Article in Special Issue
Comparative Study of Helicobacter pylori-Infected Gastritis in Okinawa and Tokyo Based on the Kyoto Classification of Gastritis
Previous Article in Journal
Uneven Meibomian Gland Dropout in Patients with Meibomian Gland Dysfunction and Demodex Infestation
Previous Article in Special Issue
Evaluation of a New Monoclonal Chemiluminescent Immunoassay Stool Antigen Test for the Diagnosis of Helicobacter pylori Infection: A Spanish Multicentre Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current and Future Perspectives in the Diagnosis and Management of Helicobacter pylori Infection

by
Malek Shatila
and
Anusha Shirwaikar Thomas
*
Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(17), 5086; https://0-doi-org.brum.beds.ac.uk/10.3390/jcm11175086
Submission received: 17 July 2022 / Revised: 24 August 2022 / Accepted: 25 August 2022 / Published: 30 August 2022

Abstract

:
Helicobacter pylori (Hp) is a prevalent organism infecting almost half the global population. It is a significant concern, given its associated risk of gastric cancer, which is the third leading cause of cancer death globally. Infection can be asymptomatic or present with dyspeptic symptoms. It may also present with alarm symptoms in the case of progression to cancer. Diagnosis can be achieved non-invasively (breath tests, stool studies, or serology) or invasively (rapid urease test, biopsy, or culture). Treatment involves acid suppression and regimens containing several antibiotics and is guided by resistance rates. Eradication is essential, as it lowers the risk of complications and progression to cancer. Follow-up after eradication is similarly important, as the risk of cancer progression remains. There have been many recent advances in both diagnosis and treatment of Hp. In particular, biosensors may be effective diagnostic tools, and nanotechnology, vaccines, and potassium-competitive acid blockers may prove effective in enhancing eradication rates.

1. Introduction

Hp is a Gram-negative, characteristically curved bacteria that was first observed to be present in the stomach lining in the 19th century [1,2]. It was not until 40 years ago that its association with gastric inflammation was demonstrated [3]. The finding that Hp could cause gastritis earned the investigators the Nobel Prize in 2005 for its myriad implications [2]. This discovery generated a massive body of research into the manifestations, treatments, and associations of Hp infection and revolutionized our understanding of the role of pathogens in disease [2,4].

Why Is H. Pylori Important?

Hp is strongly associated with duodenal ulcers (present in as many as 90% of cases), gastric ulcers (up to 80%), and malignancy; it can lead to mucosa-associated lymphoid tissue (MALT) lymphoma, as well as gastric cancer in as many as 90% of cases [5]. In 2014, the World Health Organization (WHO) called for the elimination of Hp as a means to decrease gastric cancer mortality worldwide, and in 2017. it deemed clarithromycin-resistant Hp strains a serious threat to public health [6]. In this review, we will present an overview of H. pylori disease characteristics, including epidemiology and clinical presentation, and discuss the most recent advances in evaluation and management of this entity.

2. Epidemiology

Helicobacter pylori is highly ubiquitous [7], colonizing roughly half of the world’s population [7,8]. The primary mechanism of transmission is yet to be identified but is presumed to involve person-to-person transmission [9] through fecal/oral exposure. The prevalence of Hp varies widely by region; Asia, Latin America, and Africa tend to have higher rates (up to 80% in some countries), whereas North America and Oceania have the lowest rates (as low as 24%) [10,11]; those born in the 1930s have a much higher prevalence than those born in the 1970s [10]. One meta-analysis estimated the global prevalence of Hp at 44.3%, ranging from 34.7% in developed countries to 50.8% in developing countries [12], and most recent studies show a continuous decline in Hp prevalence over the years [13]. Despite this trend, there is a worrisome increase in antibiotic-resistant Hp strains [14].

2.1. Antibiotic Resistance

Unsuccessful eradication has been a recurrent issue over the years; treatment failure rates are continuously on the rise, in large part due to the increasing prevalence of antibiotic-resistant Hp strains [14,15,16,17,18]. This is a global phenomenon affecting most countries, although specific resistance rates vary by region and antibiotic type [19]. One meta-analysis found that clarithromycin resistance reached as high as 35% in eastern Mediterranean, European, and western Pacific regions, whereas it was the lowest in Africa, the Americas, and southeast Asia, at around 15% [14]. Levofloxacin showed a somewhat similar trend, with 14% resistance in the Americas, Africa, and Europe; and around 25% in Mediterranean, southeast Asian, and western Pacific regions. Reported metronidazole rates were much higher in this study, ranging from 30 to 91%, whereas amoxicillin resistance was negligible in most regions, except for Africa, where it was 38% [14]. To put these numbers into perspective, a local resistance of >15% is the common threshold for choosing alternate treatment regimens [19,20]. In reality, the epidemiology of antibiotic resistance is far more complex, as there considerable variation within the countries of each region. This obstacle is compounded by the fact that in most regions worldwide, studies are focused primarily on a handful of countries [19]. In the US, for instance, national data are scarce, and fewer than half the states are routinely included in studies [21,22]. Better data are available from Europe, where various studies have been conducted in individual countries, as well as larger-scale projects. One study on 3974 patients by the European Registry of Helicobacter pylori Management (Hp-EuReg) found that resistance rates to clarithromycin and levofloxacin were significantly higher in southern Europe (e.g., Italy, Spain, and Greece) as opposed to northern Europe (e.g., Norway) [23]. Alarmingly, strains collected from 52% of Hp-naïve and 80% of non-naïve patients exhibited some form of antibiotic resistance in this study [23]. Antibiotic resistance may, in part, be influenced by treatment for previous Hp infections. This entity recurs in around 4% of cases annually [24,25], and various risk factors have been identified that contribute to this phenomenon.

2.2. Risk Factors

Risk factors can be categorized on a societal or individual level. The former encompasses geographic location; economic development; and sanitation, including access to clean food and water [26]. Low familial socioeconomic status and overcrowding (i.e., crowded living conditions and large family sizes) are also associated with increased Hp prevalence [26]. Consumption of unpasteurized dairy products [27], sheepherding [28], high-risk occupations (healthcare) [29], obesity [30], male gender [31], and the gut microbiome [32] pose an increased risk of infection. Smoking and alcohol are two variables that are controversial with respect to their role in Hp infections [33,34,35,36,37,38,39,40,41,42,43].

3. Etiopathogenesis

The pathogenesis of Hp infection can be divided into distinct steps, whereby the bacteria (1) attaches to and colonizes the gastric mucosa, (2) evokes and evades an immune response, and (3) induces disease. Once present in the stomach, H. pylori swims toward the mucous lining the epithelial layer, showing a tropism for sites of injury along the stomach wall [44]. This chemotaxis relies on Tlp receptors (mainly TlpB) to direct flagellar motion in response to chemical signals in the cell environment [45]. Urea, gastric acid, lactate, and reactive oxygen species have all been identified as signals for these receptors; urea in particular is secreted by the gastric epithelium and is thought to play a significant role in bacterial colonization [45]. However, undiscovered chemicals may also be implicated in this process [46]. Hp utilizes urease to protect itself from the surrounding acid environment. Urease breaks urea down into ammonia and other useful metabolites, increasing the pH in the microenvironment to create a thin, pH-neutral layer around the bacterial cell, allowing it to survive the gastric acid. This barrier reduces the viscosity of the mucin gel lining the stomach wall and allows the bacteria to move freely through the mucous toward the gastric glands that it will ultimately colonize [46,47].
Bacterial attachment to gastric epithelial cells is a complex process that involves the synergistic interaction of several elements and relies heavily on Lewis (Le) antigens. Lewis antigens are cell-surface glycoproteins that mediate cell-to-cell adhesion by binding to selectins on target cells [48]. The lipopolysaccharide (LPS) component of the Hp cell wall has been found to express Lewis-like antigens, among which LeX, in particular, has been shown to play a minor role in adhesion [49].
Bacterial outer membrane proteins (OMPs), on the other hand, operate as binding sites to which host Lewis antigens can bind, facilitating attachment. The OMPs on Hp can be divided into five genomic families [50], of which H. pylori OMP (Hop) and Helicobacter outer membrane (Hom) play the largest roles [51]. Blood antigen-binding adhesin A (BabA) and sialic acid-binding adherence (SabA) are members of the Hop family and are the most well-studied of all OMPs [52]. BabA promotes cell-to-cell adhesion by binding to host LeB [53], whereas SabA binds to sialylated LeX (sLeX) to facilitate cell adhesion [54]. SabA additionally stimulates a neutrophil response by binding neutrophil sLeX and activating a G-protein-coupled, receptor-mediated signaling cascade [55]. Interestingly, sLeX is upregulated upon the occurrence of Hp infection and gastric inflammation, which suggests that SabA may be involved in strengthening and maintaining adhesion, as opposed to initiating it [54]. MUC5a and MUC1 mucin receptors are the primary target for these OMPs and can expedite and hinder infection, respectively [56,57,58]. Whereas BabA and SabA are the main adhesins involved, several other OMPs, such as outer inflammatory protein A (OipA), HopQ, HopZ, and the Hom family, improve Hp adhesion and promote inflammation by prompting the transcription of virulence factors and the secretion of inflammatory cytokines [52].
Certain virulence factors involved in pathogenicity also contribute to adhesion. BabA-LeB binding has been shown to activate the type-four secretion system (T4SS), a pilus-like structure that allows for the translocation of effector proteins, such as cytotoxin-associated gene A (CagA) and vacuolating cytotoxin A (VacA) [54]. CagA binds to epithelial cell integrin-β1 [59] to anchor Hp and hijack host signaling pathways to disrupt cell motility, proliferation, and cytoskeletal stability [60]. VacA, on the other hand, binds to a wide range of receptors, with numerous downstream effects. It primarily functions as a pore-forming toxin than acutely induces host cell apoptosis but plays a key role in evading the immune response in chronic infections. It carries out this latter role by impairing autophagy and forming intracellular vacuoles in the host cell in which H. pylori can survive. It also binds to the integrin β2 subunit on T cells to inhibit their activation and proliferation and can induce macrophage apoptosis by inhibiting interferon-β signaling [61]. Together, these toxins are the major virulence factors expressed by Hp that are key to its pathogenicity. Whereas VacA is expressed by virtually all Hp cells, CagA is only present in specific strains; interestingly, CagA positivity is associated with more severe infection and worse clinical outcomes, including an increased risk of future malignancy [60,62]. Although these are the two major proteins involved in the pathogenesis of Hp infection, there are myriad others that similarly aid in adhesion, immune evasion, and provocation of inflammation [60].

4. Clinical Presentation

The presentation of H. pylori infection is highly variable. As many as 90% of individuals carrying the bacteria are asymptomatic [63]. It can present as dyspepsia, defined as an epigastric discomfort or pain lasting longer than one month that may be associated with nausea, early satiety, epigastric fullness, and bloating, among other symptoms [64], which is often presumed to be functional in etiology [65]. Gastrointestinal clinical symptomatology of Hp infection poorly correlates with severity of gastric mucosal injury upon endoscopy [19,64]. Hp can therefore go unnoticed and untreated, in which case it progresses to chronic gastritis [66]. This chronic inflammation of the gastric epithelium can promote intestinal metaplasia, which predisposes to gastric cancer [64]. Despite being asymptomatic for years, such patients may present with alarm symptoms of weight loss, iron deficiency anemia, dysphagia, vomiting, and the presence of an abdominal mass [19,64,65]. Similarly, MALT lymphomas may develop as a result of gastritis and present with dyspeptic or non-specific constitutional symptoms [64,67]. Hp can also cause peptic ulcer disease (PUD), with the risk of complications, such as gastrointestinal bleeding and perforation [64].
Rarely, H. pylori infection may present with extragastrointestinal manifestations, such as isolated iron deficiency anemia; idiopathic thrombocytopenic purpura; and ocular, dermatological, and metabolic diseases [65,68].

5. Evaluation and Management:

5.1. Indications for Testing

The American College of Gastroenterology (ACG) recommends testing in the following cases [65]:
(1)
All patients with active PUD;
(2)
All patients with a previous history of PUD (unless there is documentation of a resolved prior Hp infection), low-grade MALT lymphoma, or a history of endoscopic resection of early gastric cancer;
(3)
Patients with uninvestigated dyspepsia under the age of 60;
(4)
Patients initiating long-term, non-steroidal, anti-inflammatory drugs;
(5)
Patients with unexplained iron deficiency anemia despite appropriate workup; and
(6)
Adults with idiopathic thrombocytopenic purpura.
Other expert panels, including the global Taipei consensus [69] (TC) and the Houston conference [70] (HC), support guidelines similar to those outlined above, with the addition of a few other indications:
(7)
Family members residing in the same household as patients with proven active Hp infection (HC);
(8)
Patients with a family history of PUD or gastric cancer (HC);
(9)
First-generation immigrants from high-prevalence areas or high-risk groups (HC); and
(10)
Populations with a high incidence of gastric cancer (TC).
In all of the above cases, a “test-and-treat” strategy is recommended to ensure eradication of the bacteria and reduce the severity of symptoms and the risk for carcinogenesis. A variety of tests can be used to diagnose infection with H. pylori, and they can be divided into non-invasive and invasive tests, a summary of which can be found in Table 1.

5.2. Non-Invasive Tests

The most commonly used test to identify H. pylori is the urea breath test (UBT), which measures the difference in proportion of 13C/14C in exhaled air before and after the patient swallows radioactively labeled urea. This relies on the previously discussed Hp urease, which generates radiolabeled 13C carbon dioxide as a result. Patients with active Hp infection exhale higher quantities of 13C than healthy patients. Typically, four respiratory samples are collected (two before and two after ingestion of urea), and the labeled carbon dioxide is detected using mass spectrometry [71]. UBT is an accessible and commonly used tool to diagnose Hp infection, with a recent meta-analysis showing sensitivity and specificity of around 95% [72,73]. However, results can be influenced by concurrent medications, so patients are required to stop antibiotics 30 days prior to the test and proton pump inhibitors (PPIs) 15 days prior, as they may produce false negatives [71].
Stool antigen testing (SAT) is another low-cost and accurate means of diagnosing Hp infection that is often preferred by patients and physicians for its simplicity [71]. There are two main types of stool tests: enzyme immunoassays (EIAs) and immunochromatography assays (ICAs). Both tests operate under similar principles. In EIAs, a solution containing monoclonal or polyclonal antibodies is added to a diluted stool sample and processed, antigens detected through spectrometry [74]. In immunochromatography, antigen detection is made possible by a reaction caused by the antigen–antibody complex, producing a visible color change in the medium [75]. Between these two testing methods, EIA has proven to be a more effective diagnostic tool, although the accuracy of SATs varies widely depending on the detection kit used [76,77].
The final non-invasive method for diagnosing H. pylori is serological testing for immunoglobulin G (IgG) antibodies against Hp using an enzyme-linked immunosorbent assay that operates in a similar fashion to SAT. IgG antibodies appear about three weeks after infection and can remain detectable for several years thereafter. Serological tests have fallen out of favor for this reason, as they have trouble distinguishing past infections from recent or active infections and are associated with a risk of false positives [78]. Despite a high reported specificity and sensitivity for serologic antibody tests ranging from 80 to 95%, these values vary considerable depending on the testing kit used, with values reported as low as 55% [72,78]. This is especially true in the case of the newer latex agglutination immunoassay test. The latex or LZ test is increasingly used due to its low cost and rapid processing ability [79]. However, it relies on the agglutination of latex-bound antigens, and interpretation of results is highly subjective, potentially increasing the risk of false positives [79]. To their credit, serological tests are not affected by recent PPI or antibiotic use, making them an option for patients who have used either.

5.3. Invasive Tests

Endoscopic assessment is a critical component of invasive testing for H. pylori upon which all other invasive tests depend [71]. Conventional endoscopy alone is, for the most part, inadequate in diagnosing Hp infection, and a biopsy with histological assessment is still mandatory for evaluation [19]. However, advancements in endoscopic technology have allowed for image-enhanced scoping to improve the accuracy of endoscopic evaluation [19,80]. There is one scoring system currently in use called the Kyoto classification [81] that is used to evaluate active H. pylori infection and risk of gastric cancer. It consists of five endoscopic findings (atrophy, intestinal metaplasia, enlarged folds, nodularity, and diffuse redness), cumulating in a score ranging from 0 to 8. A Kyoto score ≥2 indicates H. pylori infection, whereas a score ≥4 suggests gastric cancer risk [80]. Whereas a few studies have supported the accuracy of the Kyoto classification in diagnosing active H. pylori infection [81,82], endoscopy is rarely the only diagnostic technique used to evaluate Hp infection. Instead, it is almost always paired with biopsies or alternative tests.
The gold standard for H. pylori diagnosis is histological examination. For this evaluation, at least six biopsies must be taken during biopsy targeting the antrum, large and small curvatures of the stomach, and the middle of the gastric body, as well as any suspicious lesions or ulcerations [71]. Hematoxylin–eosin and Giemsa stains are the most inexpensive and commonly used stains, but immunohistochemical staining is the most accurate (fluorescence in situ hybridization) and is recommended when histochemical methods fail [83]. The updated Sydney grading system relies on histopathological findings to assess the severity of chronic gastritis and categorizes the intensity of mononuclear inflammatory cellular infiltrates, polymorph activity, atrophy, intestinal metaplasia, and Hp density as mild, moderate, or severe [84]. The sensitivity and specificity of histological methods can range from 60% to 100% and depend on a variety of factors, including stain used; location, size, and quality of the sample; and the pathologist’s experience [71].
Alternative testing methods include bacterial culture, molecular testing (polymerase chain reaction; PCR), and rapid urease test (RUT). Bacterial culture is a highly specific means of diagnosing H. pylori infection but, as mentioned earlier, can be an arduous task that requires well-equipped laboratories [71]. Hp is a notoriously difficult micro-organism to grow and requires incubation for more than a week in selective blood agar (a detailed description of culture requirements can be found in the study by Blanchard and colleagues) [85]. It has the additional benefit of identifying antibiotic resistance, as does PCR testing, which is becoming increasingly necessary [71]. The RUT, on the other hand, relies on a pH indicator that changes color in response to the ammonia produced by Hp urease. Previous studies have shown it to be highly sensitive and specific, at ≥90%, and it has the added benefit of rapidly producing results (within 5 min with some tests). Given its ease of use, it is considered the first-line diagnostic method in cases for which endoscopy is indicated [71].
Table 1. Sensitivities and specificities of various diagnostic modalities.
Table 1. Sensitivities and specificities of various diagnostic modalities.
TestSensitivitySpecificityCost [86]Advantages [87]Disadvantages [87]Study
UBT97%96%Cost-effectiveFast, simple, non-invasive, good for detecting eradicationPotential risk for false negatives in cases of bleeding and PPI or antibiotic use; low accuracy in atrophic gastritis and gastric malignancyAbd Rahim et al., 2019 [72]
Zhou et al., 2017 [88]
Fecal antigen test94%97%Cost-effectiveFast, simple, inexpensive, can potentially be used to determine antibiotic sensitivityFalse negatives in cases of low bacterial load; accuracy affected by recent antibiotic, bismuth, or PPI use; may be uncomfortable for patients; difficulty maintaining sample; and variable accuracy depending on commercial kit usedGisbert et al., 2006 [89]
SerologyVariable (76–84%)Variable (79–90%)Cost-effectiveCheapest, widely available, can be used in patients with recent PPI or antibiotic useFailure to distinguish between acute and previous infection; cannot confirm eradicationThaker et al., 2016 [90]
Rapid Urease TestVariable (80–99%)Variable (92–100%)Cost-effectiveFast, inexpensive, simpleAccuracy impaired by gastric ulcer bleeding or intestinal metaplasia; invasiveRoy et al., 2016 [91]
Bacterial CultureVariable (70–80%)100%ExpensiveDetermination of antibiotic resistance and sensitivity Expensive, time-consuming, requires a well-equipped labThaker et al., 2016 [90]
PCR96%98%ExpensiveHigh sensitivity and specificity; effective, even at low bacterial loadsExpensive, requires a well-equipped lab, false-positive risk due to detection of DNA from dead bacteriaPichon et al., 2020 [92]

5.4. Management

Most societies endorse the non-invasive “test-and-treat” method [93,94,95], and initial endoscopy is recommended for older patients and those who present with alarm symptoms (first-degree relative with upper GI malignancy, weight loss, GI bleeding, dysphagia, odynophagia, persistent vomiting, and abnormal imaging) [94,95].
The first-line management of confirmed H. pylori infections utilizes a PPI, alongside 2–3 antibiotics for periods ranging from 3 to 14 days [64]. A full breakdown of the available first- and second-line treatment modalities can be found in Table 2.
Local antibiotic resistance rates are a key factor in determining the most appropriate first-line of management, as reflected by several major guidelines [20,64,65,96,97]. Furthermore, reuse of antibiotics from first-line treatments in subsequent therapy can lead to secondary antibiotic resistance and should be avoided [98]. Resistance to nitroimidazole antibiotics, such as metronidazole, are interesting in that whereas resistance rates can be high, this resistance can be overcome with dose adjustments and the addition of bismuth, allowing for its reuse in new regimens after an initial failure [98]. Whereas guidelines are not clear on sensitivity testing for first-line management of Hp, susceptibility testing is recommended in refractory cases [98].
An important consideration in initial management of infection is that the most commonly used acid suppressants, PPIs, are metabolized by hepatic cytochrome P450 (CYP2C19) [99]. Genetic polymorphisms may limit the efficacy of treatment regimens with particular PPIs [100]. It may be advisable to rely on PPIs less affected by CYP metabolism, such as esomeprazole and rabeprazole, especially in non-Asian regions, where extensive metabolizers are common [66,98].
The exact recommendation for treatment duration differs depending on the guidelines and treatment line (Table 2). However, there is a trend toward standardization of all treatment lines to 14 days [108].
Table 2 highlights various treatment strategies: sequential, hybrid, concomitant, and reverse hybrid. Sequential therapy involves initial dual treatment with a PPI and amoxicillin for 5–7 days, followed by standard triple therapy for the same amount of time [108]. Concomitant therapy is a non-bismuth quadruple therapy and involves the concurrent administration of four medications [109]. Hybrid therapy combines the two, initiating patients on dual therapy (PPI + amoxicillin) for 7 days, then adding two more antibiotics for the next 7 days. Reverse hybrid therapy follows the opposite order: three antibiotics and a PPI for 7 days, followed by only a PPI + amoxicillin [110]. Sequential therapy has fallen out of favor due to worse eradication rates [20,64,66,95,96,108,109]. Hybrid and reverse-hybrid treatment strategies have proven to be equivalent to concomitant across various studies but are of limited efficacy in areas of high dual resistance to clarithromycin and metronidazole [111,112]. Concomitant therapy is currently the most widely used treatment strategy.

5.5. Treatment Outcomes

The ultimate goal of treatment is documented eradication of the bacteria [20,65,96,97]. There is evidence for endoscopic and histologic remission of gastritis features, including intestinal metaplasia and reduction in recurrence following eradication therapy [69,113,114,115,116,117,118,119,120], as well as prevention of gastric adenocarcinoma and regression of gastric MALT lymphoma [121,122,123,124,125,126,127]. Treatment also resolves H. pylori-associated iron deficiency anemia [128] and ITP [129].

6. Long-Term Surveillance and Complications

6.1. Surveillance

It is currently recommended to retest at least 4 weeks after completion of the initial treatment regimen, with the patient stopping PPIs as many as 2 weeks prior [64]. All aforementioned diagnostic tests are suitable for confirmation of eradication.
Despite the lack of established evidence-based guidelines, a growing body of literature supports endoscopic surveillance following eradication, particularly in high-risk patients [130,131,132,133]. The “ABC method” relies on an investigation of anti-H. pylori antibodies and serum pepsinogen (PG), whereby patients are divided into four groups depending on the presence of either (group A, negative for both; group B, anti-Hp-positive and PG-negative; group C, positive for both; group D, anti-Hp-negative and PG-positive). Groups B, C, and D were found to be increasingly more likely to develop gastric cancer than group A and were therefore recommended triennial, biennial, and annual endoscopic follow-up based on the increased risk [134]. Nonetheless, regular endoscopy is invasive, costly, and impractical in certain settings. A recent study identified several biomarkers that could potentially be used in lieu of endoscopy for screening, detection, and monitoring of individuals at risk of gastric cancer (GC), namely virulence markers, genomic markers, transcriptomic markers, and inflammatory markers [135]. cagA and VacA-toxin expression; pepsinogen levels (PG1 and PG2); bacterial lipopolysaccharides; connexin expression; specific microRNA fragments; and cytokines, such as IL-1β, IL-6, IFN-γ, and IL-10, have been found to be significantly upregulated in cases of GC [136,137,138,139].

6.2. Complications

Treatment of H. pylori does not guarantee permanent eradication. One prospective study of 1050 patients estimated recurrence rates at one and three years to be 1.75% and 4.61%, respectively [140]. Recurrent infection necessitates alternative antibiotic treatment regimens to those used previously and may contribute to increased antibiotic resistance globally. Oral colonization of H. pylori is a potential source of reinfection that can often go undiagnosed by standard diagnostic methods for Hp and is unaffected by traditional treatment methods, requiring specific treatment strategies [141,142]. The existence of a secondary H. pylori colonization site is still highly controversial but may be a consideration in complex cases with frequent recurrences [141,142]. The main complications of untreated H. pylori infection were outlined above but include gastric ulceration; perforation; progression to gastric malignancy (adenocarcinoma or MALT lymphoma); and extragastrointestinal manifestations, such as ITP and iron deficiency anemia [65]. Other complications take the form of side effects to treatment and are listed in Table 3.

7. Treatment Challenges

H. pylori eradication rates may not be optimally attributed primarily to antibiotic resistance and patient noncompliance/adherence due to side effects and provider prescriptive error [146,147,148]. These data highlight the importance of improved awareness and strict adherence to existing guidelines.

8. Recent Advances

In the decades since its discovery, considerable progress has been made with regards to diagnostic and therapeutic modalities in the management of Hp infection. Nanotechnology is an exciting innovation in the realm of diagnosis and treatment that could eventually represent cost-efficient and less invasive alternative to current endoscopic measures. Biosensors are one such tool that can translate unique biological elements attached to a transducer surface into detectable signals [149]. This is accomplished without additional reagents, reactions, or sample pretreatment, contrary to other modalities, such as PCR or ELISA, while providing accurate and real-time monitoring of disease [149,150,151]. Whereas the process itself is complex, it relies on detecting bacterial antigens or patient Hp antibodies. Electrochemical sensors rely on a change in electric potential or conductance of a transducing surface upon element attachment, optical sensors rely on a change in fluorescence or color absorbance, piezoelectric sensors rely on a change in acoustics, and thermal sensors rely on a change in temperature to detect disease [151]. Yadav et al. (2022) mentioned the novel use of aptamers, single stranded nucleic acid sequences that are highly specific to target antigens, proteins, or antibodies, to diagnose disease and have high hopes for their clinical utility [152].
Recent advances in therapy make potential use of nanotechnology as auxiliary treatment, improving drug delivery, with a direct antibacterial effect [153]. The development of novel drugs can also improve treatment of Hp. A potassium-competitive acid blocker, vonoprazan (VPZ) has advantages over traditional acid suppression with PPIs, in that it does not require activation by gastric acid and has a longer half-life than PPIs [154]. It is also unaffected by genetic polymorphisms in CYP450 [99]. Genetic polymorphisms are receiving increasing attention for their potential role in treatment outcomes. A few studies to date have shown that eradication rates may be higher in slower CYP metabolizers [99,155,156], although this association has not always been statistically significant. Polymorphisms in immune response genes can similarly impact disease severity and predispose to complications [157,158,159]. VPZ was only recently introduced in east Asia, and several meta-analyses show the superior efficacy of VPZ relative to standard PPI-containing triple therapy [154,160,161,162]. A more recent meta-analysis of RCTs found that VPZ demonstrated comparable and even superior eradication rates relative to PPI across different treatment regimens and in both low- and high-clarithromycin-resistance areas [162]. A lower rate of adverse events was also reported among VPZ users [162].
Antimicrobial peptides are short, positively-charged peptide chains that disrupt the integrity of the negatively charged bacterial cell membrane, leading to cell lysis and disruption of intracellular processes [163]. Photodynamic therapy relies on microbial production of photosensitive molecules that use light to produce cytotoxic reactive oxygen species, leading to bacterial cell death [164]. Phage therapy uses bacteriophages specific to H. pylori to induce bacterial cell lysis, eliminating the pathogen [165]. Finally, vaccination is an attractive strategy for combatting Hp infection globally. Various attempts have been made to develop an Hp vaccine over the years, but the results have been disappointing [166]. Whereas some vaccine candidates have shown potential as an option for prophylaxis, none have yet shown a therapeutic effect [166,167].

9. Conclusions

H. pylori is a ubiquitous and complex organism that has rightfully received tremendous interest over the years. It can manifest in a variety of ways and increases the risk of severe complications, such as peptic ulceration and malignancy. Therefore, treatment with adequate follow-up is imperative. In the current era of antibiotic stewardship, it is important to be mindful of antibiotic resistance and susceptibility when selecting a treatment regimen. Extensive research has been conducted on the pathogenesis of Hp infection, which has aided in identifying diagnostic and therapeutic targets. However, there is still room to improve our knowledge. In particular, there is more to be gleaned regarding bacterial transmission, reinfection, and optimized surveillance. Finally, there have been numerous recent technological advances that hold promise for streamlining the management of this pathogen in the future.

Author Contributions

Conceptualization, M.S. and A.S.T.; writing—original draft preparation, M.S.; writing, M.S.; review and editing, A.S.T.; supervision, A.S.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bizzozero, G.; der Eidechsen, D. Ueber die schlauchförmigen Drüsen des Magendarmkanals und die Beziehungen ihres. Arch. Mikrosk. Anat. 1893, 42, 82. [Google Scholar] [CrossRef]
  2. Krienitz, W. Ueber das Auftreten von Spirochäten verschiedener form im Mageninhalt bei Carcinoma ventriculi. DMW Dtsch. Med. Wochenschr. 1906, 32, 872. [Google Scholar] [CrossRef]
  3. Watts, G. Nobel prize is awarded to doctors who discovered H. pylori. BMJ 2005, 331, 795. [Google Scholar] [CrossRef] [PubMed]
  4. Sonnenberg, A. Epidemiology of Helicobacter pylori. Aliment. Pharmacol. Ther. 2022, 55 (Suppl. 1), S1–S13. [Google Scholar] [CrossRef]
  5. Fong, I.W. Helicobacter pylori Infection: When Should It Be Treated? In Current Trends and Concerns in Infectious Diseases. Emerging Infectious Diseases of the 21st Century; Springer: Cham, Switzerland, 2020. [Google Scholar] [CrossRef]
  6. WHO. Media Centre. News Release. WHO Publishes List of Bacteria for Which New Antibiotics Are Urgently Needed. 2017. Available online: http://www.who.int/mediacentre/news/releases/2017/bacteria-antibiotics-needed/en/ (accessed on 27 August 2022).
  7. 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]
  8. Salama, N.R.; Hartung, M.L.; Müller, A. Life in the human stomach: Persistence strategies of the bacterial pathogen Helicobacter pylori. Nat. Rev. Microbiol. 2013, 11, 385–399. [Google Scholar] [CrossRef] [PubMed]
  9. Kayali, S.; Manfredi, M.; Gaiani, F.; Bianchi, L.; Bizzarri, B.; Leandro, G.; Di Mario, F.; Angelis, G.L.D. Helicobacter pylori, transmission routes and recurrence of infection: State of the art. Acta Bio Med. Atenei Parm. 2018, 89, 72–76. [Google Scholar] [CrossRef]
  10. FitzGerald, R.; Smith, S.M. An Overview of Helicobacter pylori Infection. In Helicobacter pylori. Methods in Molecular Biology; Smith, S.M., Ed.; Humana: New York, NY, USA, 2021; Volume 2283. [Google Scholar] [CrossRef]
  11. Sjomina, O.; Pavlova, J.; Niv, Y.; Leja, M. Epidemiology of Helicobacter pylori infection. Helicobacter 2018, 23 (Suppl. 1), e12514. [Google Scholar] [CrossRef] [Green Version]
  12. 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]
  13. Mezmale, L.; Coelho, L.G.; Bordin, D.; Leja, M. Review: Epidemiology of Helicobacter pylori. Helicobacter 2020, 25, e12734. [Google Scholar] [CrossRef]
  14. Savoldi, A.; Carrara, E.; Graham, D.Y.; Conti, M.; Tacconelli, E. Prevalence of Antibiotic Resistance in Helicobacter pylori: A Systematic Review and Meta-analysis in World Health Organization Regions. Gastroenterology 2018, 155, 1372–1382.e17. [Google Scholar] [CrossRef] [PubMed]
  15. Egan, B.J.; Katicic, M.; O’Connor, H.J.; O’Morain, C.A. Treatment of Helicobacter pylori. Helicobacter 2007, 12, 31–37. [Google Scholar] [CrossRef]
  16. Selgrad, M.; Meile, J.; Bornschein, J.; Kandulski, A.; Langner, C.; Varbanova, M.; Wex, T.; Tammer, I.; Schlüter, D.; Malfertheiner, P. Antibiotic susceptibility of Helicobacter pylori in central Germany and its relationship with the number of eradication therapies. Eur. J. Gastroenterol. Hepatol. 2013, 25, 1257–1260. [Google Scholar] [CrossRef] [PubMed]
  17. Zou, Y.; Qian, X.; Liu, X.; Song, Y.; Song, C.; Wu, S.; An, Y.; Yuan, R.; Wang, Y.; Xie, Y. The effect of antibiotic resistance on Helicobacter pylori eradication efficacy: A systematic review and meta-analysis. Helicobacter 2020, 25, e12714. [Google Scholar] [CrossRef]
  18. 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]
  19. Malfertheiner, P.; Megraud, F.; O’Morain, C.A.; Gisbert, J.P.; Kuipers, E.J.; Axon, A.T.; Bazzoli, F.; Gasbarrini, A.; Atherton, J.; European Helicobacter and Microbiota Study Group and Consensus panel; et al. Management of Helicobacter pylori infection—The Maastricht V/Florence Consensus Report. Gut 2017, 66, 6–30. [Google Scholar] [CrossRef]
  20. Fallone, C.A.; Chiba, N.; van Zanten, S.V.; Fischbach, L.; Gisbert, J.P.; Hunt, R.H.; Jones, N.L.; Render, C.; Leontiadis, G.I.; Moayyedi, P.; et al. The Toronto Consensus for the Treatment of Helicobacter pylori Infection in Adults. Gastroenterology 2016, 151, 51–69.e14. [Google Scholar] [CrossRef]
  21. Hulten, K.G.; Lamberth, L.B.; Kalfus, I.N.; Graham, D.Y. National and Regional US Antibiotic Resistance to Helicobacter pylori: Lessons from a Clinical Trial. Gastroenterology 2021, 161, 342–344.e1. [Google Scholar] [CrossRef]
  22. Ho, J.J.C.; Navarro, M.; Sawyer, K.; Elfanagely, Y.; Moss, S.F. Helicobacter pylori Antibiotic Resistance in the United States Between 2011–2021: A Systematic Review and Meta-Analysis. Am. J. Gastroenterol. 2022, 117, 1221–1230. [Google Scholar] [CrossRef]
  23. Bujanda, L.; Nyssen, O.P.; Vaira, D.; Saracino, I.M.; Fiorini, G.; Lerang, F.; Georgopoulos, S.; Tepes, B.; Heluwaert, F.; Gasbarrini, A.; et al. Antibiotic Resistance Prevalence and Trends in Patients Infected with Helicobacter pylori in the Period 2013–2020: Results of the European Registry on H. pylori Management (Hp-EuReg). Antibiotics 2021, 10, 1058. [Google Scholar] [CrossRef]
  24. Hu, Y.; Wan, J.-H.; Li, X.-Y.; Zhu, Y.; Graham, D.Y.; Lu, N.-H. Systematic review with meta-analysis: The global recurrence rate of Helicobacter pylori. Aliment. Pharmacol. Ther. 2017, 46, 773–779. [Google Scholar] [CrossRef]
  25. Zhao, H.; Yan, P.; Zhang, N.; Feng, L.; Chu, X.; Cui, G.; Qin, Y.; Yang, C.; Wang, S.; Yang, K. The recurrence rate of Helicobacter pylori in recent 10 years: A systematic review and meta-analysis. Helicobacter 2021, 26, e12852. [Google Scholar] [CrossRef] [PubMed]
  26. Kotilea, K.; Bontems, P.; Touati, E. Epidemiology, Diagnosis and Risk Factors of Helicobacter pylori Infection. In Helicobacter pylori in Human Diseases. Advances in Experimental Medicine and Biology; Kamiya, S., Backert, S., Eds.; Springer: Cham, Switzerland, 2019; Volume 1149. [Google Scholar] [CrossRef]
  27. Assaad, S.; Chaaban, R.; Tannous, F.; Costanian, C. Dietary habits and Helicobacter pylori infection: A cross sectional study at a Lebanese hospital. BMC Gastroenterol. 2018, 18, 48. [Google Scholar] [CrossRef] [PubMed]
  28. Dore, M.P.; Bilotta, M.; Vaira, D.; Manca, 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]
  29. Mastromarino, P.; Conti, C.; Donato, K.; Strappini, P.; Cattaruzza, M.; Orsi, G. Does hospital work constitute a risk factor for Helicobacter pylori infection? J. Hosp. Infect. 2005, 60, 261–268. [Google Scholar] [CrossRef]
  30. Baradaran, A.; Dehghanbanadaki, H.; Naderpour, S.; Pirkashani, L.M.; Rajabi, A.; Rashti, R.; Riahifar, S.; Moradi, Y. The association between Helicobacter pylori and obesity: A systematic review and meta-analysis of case–control studies. Clin. Diabetes Endocrinol. 2021, 7, 15. [Google Scholar] [CrossRef] [PubMed]
  31. de Martel, C.; Parsonnet, J. Helicobacter pylori Infection and Gender: A Meta-Analysis of Population-Based Prevalence Surveys. Am. J. Dig. Dis. 2006, 51, 2292–2301. [Google Scholar] [CrossRef] [PubMed]
  32. Wang, D.; Li, Y.; Zhong, H.; Ding, Q.; Lin, Y.; Tang, S.; Zong, Y.; Wang, Q.; Zhang, X.; Yang, H.; et al. Alterations in the human gut microbiome associated with Helicobacter pylori infection. FEBS Open Bio. 2019, 9, 1552–1560. [Google Scholar] [CrossRef]
  33. Wang, W.; Jiang, W.; Zhu, S.; Sun, X.; Li, P.; Liu, K.; Liu, H.; Gu, J.; Zhang, S. Assessment of prevalence and risk factors of helicobacter pylori infection in an oilfield Community in Hebei, China. BMC Gastroenterol. 2019, 19, 186. [Google Scholar] [CrossRef]
  34. Zhang, F.; Pu, K.; Wu, Z.; Zhang, Z.; Liu, X.; Chen, Z.; Ye, Y.; Wang, Y.; Zheng, Y.; Zhang, J.; et al. Prevalence and associated risk factors of Helicobacter pylori infection in the Wuwei cohort of north-western China. Trop. Med. Int. Health 2021, 26, 290–300. [Google Scholar] [CrossRef]
  35. Zhu, Y.; Zhou, X.; Wu, J.; Su, J.; Zhang, G. Risk Factors and Prevalence of Helicobacter pylori Infection in Persistent High Incidence Area of Gastric Carcinoma in Yangzhong City. Gastroenterol. Res. Pract. 2014, 2014, 481365. [Google Scholar] [CrossRef] [PubMed]
  36. Smith, S.; Jolaiya, T.; Fowora, M.; Palamides, P.; Ngoka, F.; Bamidele, M.; Lesi, O.; Onyekwere, C.; Ugiagbe, R.; Agbo, I.; et al. Clinical and Socio-Demographic Risk Factors for Acquisition of Helicobacter pylori Infection in Nigeria. Asian Pac. J. Cancer Prev. 2018, 19, 1851–1857. [Google Scholar] [CrossRef]
  37. Zhang, L.; Eslick, G.D.; Xia, H.H.-X.; Wu, C.; Phung, N.; Talley, N.J. Relationship between Alcohol Consumption and Active Helicobacter pylori Infection. Alcohol Alcohol. 2010, 45, 89–94. [Google Scholar] [CrossRef] [PubMed]
  38. Seid, A.; Demsiss, W. Feco-prevalence and risk factors of Helicobacter pylori infection among symptomatic patients at Dessie Referral Hospital, Ethiopia. BMC Infect. Dis. 2018, 18, 260. [Google Scholar] [CrossRef] [PubMed]
  39. Wu, W.; Leja, M.; Tsukanov, V.; Basharat, Z.; Hua, D.; Hong, W. Sex differences in the relationship among alcohol, smoking, and Helicobacter pylori infection in asymptomatic individuals. J. Int. Med. Res. 2020, 48. [Google Scholar] [CrossRef] [PubMed]
  40. Ogihara, A.; Kikuchi, S.; Hasegawa, A.; Kurosawa, M.; Miki, K.; Kaneko, E.; Mizukoshi, H. Relationship between Helicobacter pylori infection and smoking and drinking habits. J. Gastroenterol. Hepatol. 2000, 15, 271–276. [Google Scholar] [CrossRef]
  41. Ozaydin, N.; A Turkyilmaz, S.; Cali, S. Prevalence and risk factors of helicobacter pylori in Turkey: A nationally-representative, cross-sectional, screening with the 13C-Urea breath test. BMC Public Health 2013, 13, 1215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. de Jésus Ngoma, P.; Longo-Mbenza, B.; Tshibangu-Kabamba, E.; Matungo, B.; Lupande, D.; Malu, C.; Kengibe, P.; Yaba, A.T.W.; Nlombi, C.M.; Fiasse, R.; et al. Seroprevalence and Risk Factors of the Helicobacter pylori Infection in Bukavu City in the Democratic Republic of Congo. OALib 2021, 8, e8032. [Google Scholar] [CrossRef]
  43. Sirkeci, Ö.; Sirkeci, E.E.; Ulas, T. Does waterpipe smoking increase the risk of Helicobacter pylori infection? J. Res. Med. Sci. 2022, 27, 7. [Google Scholar] [CrossRef]
  44. Denic, M.; Touati, E.; De Reuse, H. Review: Pathogenesis of Helicobacter pylori infection. Helicobacter 2020, 25, e12736. [Google Scholar] [CrossRef]
  45. Hanyu, H.; Engevik, K.A.; Matthis, A.L.; Ottemann, K.M.; Montrose, M.H.; Aihara, E. Helicobacter pylori Uses the TlpB Receptor to Sense Sites of Gastric Injury. Infect. Immun. 2019, 87, e00202-19. [Google Scholar] [CrossRef] [PubMed]
  46. Idowu, S.; Bertrand, P.P.; Walduck, A.K. Gastric organoids: Advancing the study of H. pylori pathogenesis and inflammation. Helicobacter 2022, 27, e12891. [Google Scholar] [CrossRef] [PubMed]
  47. Celli, J.P.; Turner, B.S.; Afdhal, N.H.; Keates, S.; Ghiran, I.; Kelly, C.P.; Ewoldt, R.H.; McKinley, G.H.; So, P.; Erramilli, S.; et al. Helicobacter pylori moves through mucus by reducing mucin viscoelasticity. Proc. Natl. Acad. Sci. USA 2009, 106, 14321–14326. [Google Scholar] [CrossRef]
  48. Soejima, M.; Koda, Y. Molecular mechanisms of Lewis antigen expression. Leg. Med. 2005, 7, 266–269. [Google Scholar] [CrossRef]
  49. Odenbreit, S.; Faller, G.; Haas, R. Role of the AlpAB proteins and lipopolysaccharide in adhesion of Helicobacter pylori to human gastric tissue. Int. Med. Microbiol. 2002, 292, 247–256. [Google Scholar] [CrossRef] [PubMed]
  50. Alm, R.A.; Bina, J.; Andrews, B.M.; Doig, P.; Hancock, R.E.W.; Trust, T.J. Comparative Genomics of Helicobacter pylori: Analysis of the Outer Membrane Protein Families. Infect. Immun. 2000, 68, 4155–4168. [Google Scholar] [CrossRef]
  51. Oleastro, M.; Ménard, A. The Role of Helicobacter pylori Outer Membrane Proteins in Adherence and Pathogenesis. Biology 2013, 2, 1110–1134. [Google Scholar] [CrossRef] [Green Version]
  52. Xu, C.; Soyfoo, D.M.; Wu, Y.; Xu, S. Virulence of Helicobacter pylori outer membrane proteins: An updated review. Eur. J. Clin. Microbiol. Infect. Dis. 2020, 39, 1821–1830. [Google Scholar] [CrossRef]
  53. Hage, N.; Howard, T.; Phillips, C.; Brassington, C.; Overman, R.; Debreczeni, J.; Gellert, P.; Stolnik, S.; Winkler, G.S.; Falcone, F.H. Structural basis of Lewis b antigen binding by the Helicobacter pylori adhesin BabA. Sci. Adv. 2015, 1, e1500315. [Google Scholar] [CrossRef]
  54. Doohan, D.; Rezkitha, Y.A.A.; Waskito, L.A.; Yamaoka, Y.; Miftahussurur, M. Helicobacter pylori BabA–SabA Key Roles in the Adherence Phase: The Synergic Mechanism for Successful Colonization and Disease Development. Toxins 2021, 13, 485. [Google Scholar] [CrossRef]
  55. Unemo, M.; Aspholm-Hurtig, M.; Ilver, D.; Bergström, J.; Borén, T.; Danielsson, D.; Teneberg, S. The Sialic Acid Binding SabA Adhesin of Helicobacter pylori Is Essential for Nonopsonic Activation of Human Neutrophils. J. Biol. Chem. 2005, 280, 15390–15397. [Google Scholar] [CrossRef] [PubMed]
  56. Giraldi, L.; Michelazzo, M.B.; Arzani, D.; Persiani, R.; Pastorino, R.; Boccia, S. MUC1, MUC5AC, and MUC6 polymorphisms, Helicobacter pylori infection, and gastric cancer: A systematic review and meta-analysis. Eur. J. Cancer Prevention 2018, 27, 323–330. [Google Scholar] [CrossRef] [PubMed]
  57. van de Bovenkamp, J.H.B.; Mahdavi, J.; Male, A.M.K.-V.; Buller, H.A.; Einerhand, A.W.C.; Boren, T.; Dekker, J. The MUC5AC Glycoprotein is the Primary Receptor for Helicobacter pylori in the Human Stomach. Helicobacter 2003, 8, 521–532. [Google Scholar] [CrossRef] [PubMed]
  58. Lindén, S.K.; Sheng, Y.H.; Every, A.L.; Miles, K.M.; Skoog, E.C.; Florin, T.H.J.; Sutton, P.; McGuckin, M.A. MUC1 Limits Helicobacter pylori Infection both by Steric Hindrance and by Acting as a Releasable Decoy. PLoS Pathog. 2009, 5, e1000617. [Google Scholar] [CrossRef]
  59. Backert, S.; Tegtmeyer, N. Type IV Secretion and Signal Transduction of Helicobacter pylori CagA through Interactions with Host Cell Receptors. Toxins 2017, 9, 115. [Google Scholar] [CrossRef]
  60. Baj, J.; Forma, A.; Sitarz, M.; Portincasa, P.; Garruti, G.; Krasowska, D.; Maciejewski, R. Helicobacter pylori Virulence Factors—Mechanisms of Bacterial Pathogenicity in the Gastric Microenvironment. Cells 2021, 10, 27. [Google Scholar] [CrossRef]
  61. Chauhan, N.; Tay, A.C.Y.; Marshall, B.J.; Jain, U. Helicobacter pylori VacA, a distinct toxin exerts diverse functionalities in numerous cells: An overview. Helicobacter 2019, 24, e12544. [Google Scholar] [CrossRef] [Green Version]
  62. Sharndama, H.C.; Mba, I.E. Helicobacter pylori: An up-to-date overview on the virulence and pathogenesis mechanisms. Braz. J. Microbiol. 2022, 53, 33–50. [Google Scholar] [CrossRef]
  63. Alexander, S.M.; Retnakumar, R.J.; Chouhan, D.; Devi, T.N.B.; Dharmaseelan, S.; Devadas, K.; Thapa, N.; Tamang, J.P.; Lamtha, S.C.; Chattopadhyay, S. Helicobacter pylori in Human Stomach: The Inconsistencies in Clinical Outcomes and the Probable Causes. Front. Microbiol. 2021, 12, 713955. [Google Scholar] [CrossRef]
  64. Guevara, B.; Cogdill, A.G. Helicobacter pylori: A Review of Current Diagnostic and Management Strategies. Am. J. Dig. Dis. 2020, 65, 1917–1931. [Google Scholar] [CrossRef]
  65. Chey, W.D.; Leontiadis, G.I.; Howden, C.W.; Moss, S.F. ACG Clinical Guideline: Treatment of Helicobacter pylori Infection. Am. J. Gastroenterol. 2017, 112, 212–239. [Google Scholar] [CrossRef] [PubMed]
  66. Sugano, K.; Tack, J.; Kuipers, E.J.; Graham, D.Y.; El-Omar, E.M.; Miura, S.; Haruma, K.; Asaka, M.; Uemura, N.; Kyoto Global Consensus Conference; et al. Kyoto global consensus report on Helicobacter pylori gastritis. Gut 2015, 64, 1353–1367. [Google Scholar] [CrossRef] [PubMed]
  67. Fischbach, W. Gastric Mucosal-Associated Lymphoid Tissue Lymphoma. Gastroenterol. Clin. N. Am. 2013, 42, 371–380. [Google Scholar] [CrossRef] [PubMed]
  68. Gravina, A.G.; Priadko, K.; Ciamarra, P.; Granata, L.; Facchiano, A.; Miranda, A.; Dallio, M.; Federico, A.; Romano, M. Extra-Gastric Manifestations of Helicobacter pylori Infection. J. Clin. Med. 2020, 9, 3887. [Google Scholar] [CrossRef]
  69. Liou, J.-M.; Malfertheiner, P.; Lee, Y.-C.; Sheu, B.-S.; Sugano, K.; Cheng, H.-C.; Yeoh, K.-G.; Hsu, P.-I.; Goh, K.-L.; Mahachai, V.; et al. Screening and eradication of Helicobacter pylori for gastric cancer prevention: The Taipei global consensus. Gut 2020, 69, 2093–2112. [Google Scholar] [CrossRef]
  70. El-Serag, H.B.; Kao, J.Y.; Kanwal, F.; Gilger, M.; LoVecchio, F.; Moss, S.F.; Crowe, S.; Elfant, A.; Haas, T.; Hapke, R.J.; et al. Houston Consensus Conference on Testing for Helicobacter pylori Infection in the United States. Clin. Gastroenterol. Hepatol. 2018, 16, 992–1002.e6, Correction in Clin. Gastroenterol. Hepatol. 2019, 17, 801. [Google Scholar] [CrossRef]
  71. Cardos, A.I.; Maghiar, A.; Zaha, D.C.; Pop, O.; Fritea, L.; Miere , F.; Cavalu, S. Evolution of Diagnostic Methods for Helicobacter pylori Infections: From Traditional Tests to High Technology, Advanced Sensitivity and Discrimination Tools. Diagnostics 2022, 12, 508. [Google Scholar] [CrossRef]
  72. Rahim, M.A.A.; Johani, F.H.; Shah, S.A.; Hassan, M.R.; Manaf, M.R.A. 13C-Urea Breath Test Accuracy for Helicobacter pylori Infection in the Asian Population: A Meta-Analysis. Ann. Glob. Health 2019, 85, 110. [Google Scholar] [CrossRef]
  73. O’Connor, A. The Urea Breath Test for the Noninvasive Detection of Helicobacter pylori. In Helicobacter Pylori. Methods in Molecular Biology; Smith, S.M., Ed.; Humana: New York, NY, USA, 2021; Volume 2283. [Google Scholar] [CrossRef]
  74. Koletzko, S.; Konstantopoulos, N.; Bosman, D.; Feydt-Schmidt, A.; van der Ende, A.; Kalach, N.; Raymond, J.; Rüssmann, H. Evaluation of a novel monoclonal enzyme immunoassay for detection of Helicobacter pylori antigen in stool from children. Gut 2003, 52, 804–806. [Google Scholar] [CrossRef]
  75. Gatta, L.; Perna, F.; Ricci, C.; Osborn, J.F.; Tampieri, A.; Bernabucci, V.; Miglioli, M.; Vaira, D. A rapid immunochromatographic assay for Helicobacter pylori in stool before and after treatment. Aliment. Pharmacol. Ther. 2004, 20, 469–474. [Google Scholar] [CrossRef]
  76. Qiu, E.; Li, Z.; Han, S. Methods for detection of Helicobacter pylori from stool sample: Current options and developments. Braz. J. Microbiol. 2021, 52, 2057–2062. [Google Scholar] [CrossRef] [PubMed]
  77. Shimoyama, T. Stool antigen tests for the management of Helicobacter pylori infection. World J. Gastroenterol. 2013, 19, 8188–8191. [Google Scholar] [CrossRef] [PubMed]
  78. Dore, M.P.; Graham, D.Y. Modern approach to the diagnosis of Helicobacter pylori infection. Aliment. Pharmacol. Ther. 2022, 55, S14–S21. [Google Scholar] [CrossRef] [PubMed]
  79. Stefano, K.; Rosalia, A.; Chiara, B.; Federica, G.; Marco, M.; Gioacchino, L.; Fabiola, F.; Francesco, D.M.; Gian, L.D.A. Non-invasive tests for the diagnosis of helicobacter pylori: State of the art. Acta Biomed. 2018, 89 (Suppl. 8), 58–64. [Google Scholar] [CrossRef]
  80. Toyoshima, O.; Nishizawa, T.; Koike, K. Endoscopic Kyoto classification of Helicobacter pylori infection and gastric cancer risk diagnosis. World J. Gastroenterol. 2020, 26, 466–477. [Google Scholar] [CrossRef]
  81. Toyoshima, O.; Nishizawa, T.; Yoshida, S.; Matsuno, T.; Odawara, N.; Toyoshima, A.; Sakitani, K.; Watanabe, H.; Fujishiro, M.; Suzuki, H. Consistency between the endoscopic Kyoto classification and pathological updated Sydney system for gastritis: A cross-sectional study. J. Gastroenterol. Hepatol. 2022, 37, 291–300. [Google Scholar] [CrossRef]
  82. Zhao, J.; Xu, S.; Gao, Y.; Lei, Y.; Zou, B.; Zhou, M.; Chang, D.; Dong, L.; Qin, B. Accuracy of Endoscopic Diagnosis of Helicobacter pylori Based on the Kyoto Classification of Gastritis: A Multicenter Study. Front. Oncol. 2020, 10, 599218. [Google Scholar] [CrossRef]
  83. White, J.R.; Sami, S.S.; Reddiar, D.; Mannath, J.; Ortiz-Fernández-Sordo, J.; Beg, S.; Scott, R.; Thiagarajan, P.; Ahmad, S.; Parra-Blanco, A.; et al. Narrow Band Imaging and Serology in the Assessment of Premalignant Gastric Pathology. Scand. J. Gastroenterol. 2018, 53, 1611–1618. [Google Scholar] [CrossRef]
  84. Hassan, T.M.M.; Al-Najjar, S.I.; Al-Zahrani, I.H.; Alanazi, F.I.B.; Alotibi, M.G. Helicobacter pylori chronic gastritis updated Sydney grading in relation to endoscopic findings and H. pylori IgG antibody: Diagnostic methods. J. Microsc. Ultrastruct. 2016, 4, 167–174. [Google Scholar] [CrossRef]
  85. Blanchard, T.G.; Nedrud, J.G. Laboratory Maintenance of Helicobacter Species. Curr. Protoc. Microbiol. 2006, 8B.1.1–8B.1.13. [Google Scholar] [CrossRef]
  86. Sulo, P.; Šipková, B. DNA diagnostics for reliable and universal identification of Helicobacter pylori. World J. Gastroenterol. 2021, 27, 7100–7112. [Google Scholar] [CrossRef] [PubMed]
  87. Sabbagh, P.; Mohammadnia-Afrouzi, M.; Javanian, M.; Babazadeh, A.; Koppolu, V.; Vasigala, V.R.; Nouri, H.R.; Ebrahimpour, S. Diagnostic methods for Helicobacter pylori infection: Ideals, options, and limitations. Eur. J. Clin. Microbiol. 2019, 38, 55–66. [Google Scholar] [CrossRef]
  88. Zhou, Q.; Li, L.; Ai, Y.; Pan, Z.; Guo, M.; Han, J. Diagnostic accuracy of the 14C-urea breath test in Helicobacter pylori infections: A meta-analysis. Wien Klin Wochenschr 2017, 129, 38–45. [Google Scholar] [CrossRef] [PubMed]
  89. Gisbert, J.P.; de la Morena, F.; Abraira, V. Accuracy of monoclonal stool antigen test for the diagnosis of H. pylori infection: A systematic review and meta-analysis. Am. J. Gastroenterol. 2006, 101, 1921. [Google Scholar] [CrossRef] [PubMed]
  90. Thaker, Y.; Moon, A.; Afzali, A. Helicobacter pylori: A review of epidemiology, treatment, and management. J. Clin. Gastroenterol. Treat. 2016, 2, 1–5. [Google Scholar] [CrossRef]
  91. Roy, A.D.; Deuri, S.; Dutta, U.C. The diagnostic accuracy of rapid urease biopsy test compared to histopathology in implementing “test and treat” policy for Helicobacter pylori. Int. J. Appl. Basic Med. Res. 2016, 6, 18–22. [Google Scholar] [CrossRef] [Green Version]
  92. Pichon, M.; Pichard, B.; Barrioz, T.; Plouzeau, C.; Croquet, V.; Fotsing, G.; Chéron, A.; Vuillemin, É.; Wangermez, M.; Haineaux, P.-A.; et al. Diagnostic Accuracy of a Noninvasive Test for Detection of Helicobacter pylori and Resistance to Clarithromycin in Stool by the Amplidiag H. pylori + ClariR Real-Time PCR Assay. J. Clin. Microbiol. 2020, 58, e01787-19. [Google Scholar] [CrossRef]
  93. Ford, A.C.; Qume, M.; Moayyedi, P.; Arents, N.L.; Lassen, A.T.; Logan, R.F.; McColl, K.E.; Myres, P.; Delaney, B.C. Helicobacter pylori “Test and Treat” or Endoscopy for Managing Dyspepsia: An Individual Patient Data Meta-analysis. Gastroenterology 2005, 128, 1838–1844. [Google Scholar] [CrossRef]
  94. ASGE Standards of Practice Committee; Shaukat, A.; Wang, A.; Acosta, R.D.; Bruining, D.H.; Chandrasekhara, V.; Chathadi, K.V.; Eloubeidi, M.A.; Fanelli, R.D.; Faulx, A.L.; et al. The role of endoscopy in dyspepsia. Gastrointest. Endosc. 2015, 82, 227–232. [Google Scholar] [CrossRef]
  95. Moayyedi, P.M.; E Lacy, B.; Andrews, C.N.; Enns, R.A.; Howden, C.W.; Vakil, N. ACG and CAG Clinical Guideline: Management of Dyspepsia. Am. J. Gastroenterol. 2017, 112, 988–1013. [Google Scholar] [CrossRef]
  96. Liu, W.Z.; Xie, Y.; Lu, H.; Cheng, H.; Zeng, Z.R.; Zhou, L.Y.; Chen, Y.; Bin Wang, J.; Du, Y.Q.; Lu, N.H.; et al. Fifth Chinese National Consensus Report on the management of Helicobacter pylori infection. Helicobacter 2018, 23, e12475. [Google Scholar] [CrossRef] [PubMed]
  97. Gisbert, J.P.; Alcedo, J.; Amador, J.; Bujanda, L.; Calvet, X.; Castro-Fernández, M.; Fernández-Salazar, L.; Gené, E.; Lanas, Á.; Lucendo, A.J.; et al. V Conferencia Española de Consenso sobre el tratamiento de la infección por Helicobacter pylori. Gastroenterol. Hepatol. 2022, 45, 392–417. [Google Scholar] [CrossRef] [PubMed]
  98. Shah, S.C.; Iyer, P.G.; Moss, S.F. AGA Clinical Practice Update on the Management of Refractory Helicobacter pylori Infection: Expert Review. Gastroenterology 2021, 160, 1831–1841. [Google Scholar] [CrossRef] [PubMed]
  99. Argueta, E.A.; Moss, S.F. How We Approach Difficult to Eradicate Helicobacter pylori. Gastroenterology 2022, 162, 32–37. [Google Scholar] [CrossRef] [PubMed]
  100. Kuo, C.-H.; Lu, C.-Y.; Shih, H.-Y.; Liu, C.-J.; Wu, M.-C.; Hu, H.-M.; Hsu, W.-H.; Yu, F.-J.; Wu, D.-C.; Kuo, F.-C. CYP2C19 polymorphism influences Helicobacter pylori eradication. World J. Gastroenterol. 2014, 20, 16029–16036. [Google Scholar] [CrossRef]
  101. Suzuki, H.; Mori, H. World trends for H. pylori eradication therapy and gastric cancer prevention strategy by H. pylori test-and-treat. J. Gastroenterol. 2018, 53, 354–361. [Google Scholar] [CrossRef] [Green Version]
  102. Murakami, K.; Sakurai, Y.; Shiino, M.; Funao, N.; Nishimura, A.; Asaka, M. Vonoprazan, a novel potassium-competitive acid blocker, as a component of first-line and second-line triple therapy for Helicobacter pylori eradication: A phase III, randomised, double-blind study. Gut 2016, 65, 1439–1446. [Google Scholar] [CrossRef]
  103. Lu, H.; Zhang, W.; Graham, D.Y. Bismuth-containing quadruple therapy for Helicobacter pylori: Lessons from China. Eur. J. Gastroenterol. Hepatol. 2013, 25, 1134–1140. [Google Scholar] [CrossRef]
  104. Hassan, A.M.; Shawky, M.A.E.-G.; Mohammed, A.Q.; Haridy, M.A.; Eid, K.A.-E. Simvastatin improves the eradication rate of Helicobacter pylori: Upper Egypt experience. Infect. Drug Resist. 2019, 12, 1529–1534. [Google Scholar] [CrossRef]
  105. Sarkeshikian, S.S.; Ghadir, M.R.; Alemi, F.; Jalali, S.M.; Hormati, A.; Mohammadbeigi, A. Atorvastatin in combination with conventional antimicrobial treatment of Helicobacter pylori eradication: A randomized controlled clinical trial. J. Gastroenterol. Hepatol. 2020, 35, 71–75. [Google Scholar] [CrossRef]
  106. Liao, W.-C.; Huang, M.-Z.; Wang, M.L.; Lin, C.-J.; Lu, T.-L.; Lo, H.-R.; Pan, Y.-J.; Sun, Y.-C.; Kao, M.-C.; Lim, H.-J.; et al. Statin Decreases Helicobacter pylori Burden in Macrophages by Promoting Autophagy. Front. Cell. Infect. Microbiol. 2017, 6, 203. [Google Scholar] [CrossRef] [PubMed]
  107. Viazis, N.; Argyriou, K.; Kotzampassi, K.; Christodoulou, D.K.; Apostolopoulos, P.; Georgopoulos, S.D.; Liatsos, C.; Giouleme, O.; Koustenis, K.; Veretanos, C.; et al. A Four-Probiotics Regimen Combined with A Standard Helicobacter pylori-Eradication Treatment Reduces Side Effects and Increases Eradication Rates. Nutrients 2022, 14, 632. [Google Scholar] [CrossRef] [PubMed]
  108. Graham, D.Y.; Liou, J.-M. Primer for Development of Guidelines for Helicobacter pylori Therapy Using Antimicrobial Stewardship. Clin. Gastroenterol. Hepatol. 2022, 20, 973–983.e1. [Google Scholar] [CrossRef] [PubMed]
  109. Wang, Y.; Zhao, R.; Wang, B.; Zhao, Q.; Li, Z.; Zhu-Ge, L.; Yin, W.; Xie, Y. Sequential versus concomitant therapy for treatment of Helicobacter pylori infection: An updated systematic review and meta-analysis. Eur. J. Clin. Pharmacol. 2018, 74, 1–13. [Google Scholar] [CrossRef] [PubMed]
  110. Georgopoulos, S.; Papastergiou, V. An update on current and advancing pharmacotherapy options for the treatment of H. pylori infection. Expert Opin. Pharmacother. 2021, 22, 729–741. [Google Scholar] [CrossRef]
  111. Du, L.-J.; Chen, B.-R.; Kim, J.J.; Kim, S.; Shen, J.-H.; Dai, N. Helicobacter pylori eradication therapy for functional dyspepsia: Systematic review and meta-analysis. World J. Gastroenterol. 2016, 22, 3486–3495. [Google Scholar] [CrossRef]
  112. Hsu, P.; Tsay, F.; Kao, J.Y.; Peng, N.; Tsai, K.; Tsai, T.; Kuo, C.; Kao, S.; Wang, H.; Chen, Y.; et al. Equivalent efficacies of reverse hybrid and concomitant therapies in first-line treatment of Helicobacter pylori infection. J. Gastroenterol. Hepatol. 2020, 35, 1731–1737. [Google Scholar] [CrossRef]
  113. Zhao, B.; Zhao, J.; Cheng, W.-F.; Shi, W.-J.; Liu, W.; Pan, X.-L.; Zhang, G.-X. Efficacy of Helicobacter pylori Eradication Therapy on Functional Dyspepsia. J. Clin. Gastroenterol. 2014, 48, 241–247. [Google Scholar] [CrossRef]
  114. Kim, S.E.; Park, Y.S.; Kim, N.; Kim, M.S.; Jo, H.J.; Shin, C.M.; Lee, S.H.; Hwang, J.-H.; Kim, J.-W.; Jeong, S.-H.; et al. Effect of Helicobacter pylori Eradication on Functional Dyspepsia. J. Neurogastroenterol. Motil. 2013, 19, 233–243. [Google Scholar] [CrossRef]
  115. Harvey, R.F.; Lane, A.; Nair, P.; Egger, M.; Harvey, I.; Donovan, J.; Murray, L. Clinical trial: Prolonged beneficial effect of Helicobacter pylori eradication on dyspepsia consultations—The Bristol Helicobacter Project. Aliment. Pharmacol. Ther. 2010, 32, 394–400. [Google Scholar] [CrossRef]
  116. Ford, A.C.; Tsipotis, E.; Yuan, Y.; I Leontiadis, G.; Moayyedi, P. Efficacy of Helicobacter pylori eradication therapy for functional dyspepsia: Updated systematic review and meta-analysis. Gut 2021, 2021, 326583. [Google Scholar] [CrossRef] [PubMed]
  117. Hwang, Y.J.; Choi, Y.; Kim, N.; Lee, H.S.; Yoon, H.; Shin, C.M.; Park, Y.S.; Lee, D.H. The Difference of Endoscopic and Histologic Improvements of Atrophic Gastritis and Intestinal Metaplasia After Helicobacter pylori Eradication. Am. J. Dig. Dis. 2021, 67, 3055–3066. [Google Scholar] [CrossRef] [PubMed]
  118. Toyoshima, O.; Nishizawa, T.; Sakitani, K.; Yamakawa, T.; Takahashi, Y.; Kinoshita, K.; Torii, A.; Yamada, A.; Suzuki, H.; Koike, K. Helicobacter pylori eradication improved the Kyoto classification score on endoscopy. JGH Open 2020, 4, 909–914. [Google Scholar] [CrossRef]
  119. Hwang, Y.-J.; Kim, N.; Lee, H.S.; Lee, J.B.; Choi, Y.J.; Yoon, H.; Shin, C.M.; Park, Y.S.; Lee, D.H. Reversibility of atrophic gastritis and intestinal metaplasia after Helicobacter pylori eradication—A prospective study for up to 10 years. Aliment. Pharmacol. Ther. 2018, 47, 380–390. [Google Scholar] [CrossRef] [PubMed]
  120. Ford, A.C.; Gurusamy, K.S.; Delaney, B.; Forman, D.; Moayyedi, P. Eradication therapy for peptic ulcer disease in Helicobacter pylori-positive people. Cochrane Database Syst. Rev. 2016, 4, CD003840. [Google Scholar] [CrossRef]
  121. Lee, Y.-C.; Chiang, T.-H.; Chou, C.-K.; Tu, Y.-K.; Liao, W.-C.; Wu, M.-S.; Graham, D.Y. Association Between Helicobacter pylori Eradication and Gastric Cancer Incidence: A Systematic Review and Meta-analysis. Gastroenterology 2016, 150, 1113–1124.e5. [Google Scholar] [CrossRef] [Green Version]
  122. Tsukamoto, T.; Nakagawa, M.; Kiriyama, Y.; Toyoda, T.; Cao, X. Prevention of Gastric Cancer: Eradication of Helicobacter pylori and Beyond. Int. J. Mol. Sci. 2017, 18, 1699. [Google Scholar] [CrossRef]
  123. Ford, A.C.; Yuan, Y.; Moayyedi, P. Helicobacter pylori eradication therapy to prevent gastric cancer: Systematic review and meta-analysis. Gut 2020, 69, 2113–2121. [Google Scholar] [CrossRef]
  124. Doorakkers, E.; Lagergren, J.; Engstrand, L.; Brusselaers, N. Eradication of Helicobacter pylori and Gastric Cancer: A Systematic Review and Meta-analysis of Cohort Studies. J. Natl. Cancer Inst. 2016, 108, djw132. [Google Scholar] [CrossRef]
  125. Nakamura, S.; Sugiyama, T.; Matsumoto, T.; Iijima, K.; Ono, S.; Tajika, M.; Tari, A.; Kitadai, Y.; Matsumoto, H.; Nagaya, T.; et al. Long-term clinical outcome of gastric MALT lymphoma after eradication of Helicobacter pylori: A multicentre cohort follow-up study of 420 patients in Japan. Gut 2012, 61, 507–513. [Google Scholar] [CrossRef]
  126. Hu, Q.; Zhang, Y.; Zhang, X.; Fu, K. Gastric mucosa-associated lymphoid tissue lymphoma and Helicobacter pylori infection: A review of current diagnosis and management. Biomark. Res. 2016, 4, 15. [Google Scholar] [CrossRef] [PubMed]
  127. Filip, P.V.; Cuciureanu, D.; Diaconu, L.S.; Vladareanu, A.M.; Pop, C.S. MALT lymphoma: Epidemiology, clinical diagnosis and treatment. J. Med. Life 2018, 11, 187–193. [Google Scholar] [CrossRef] [PubMed]
  128. Kim, B.J.; Kim, H.S.; Jang, H.J.; Kim, J.H. Helicobacter pylori Eradication in Idiopathic Thrombocytopenic Purpura: A Meta-Analysis of Randomized Trials. Gastroenterol. Res. Pract. 2018, 2018, 6090878. [Google Scholar] [CrossRef] [PubMed]
  129. Sapmaz, F.; Basyigit, S.; Kısa, Ü.; Kavak, E.E.; Guliter, S.; Kalkan, I.H. The impact of Helicobacter pylori eradication on serum hepcidin-25 level and iron parameters in patients with iron deficiency anemia. Wien. Klin. Wochenschr. 2016, 128, 335–340. [Google Scholar] [CrossRef] [PubMed]
  130. Kishikawa, H.; Ojiro, K.; Nakamura, K.; Katayama, T.; Arahata, K.; Takarabe, S.; Miura, S.; Kanai, T.; Nishida, J. Previous Helicobacter pylori infection-induced atrophic gastritis: A distinct disease entity in an understudied population without a history of eradication. Helicobacter 2020, 25, e12669. [Google Scholar] [CrossRef]
  131. Shichijo, S.; Hirata, Y. Characteristics and predictors of gastric cancer after Helicobacter pylori eradication. World J. Gastroenterol. 2018, 24, 2163–2172. [Google Scholar] [CrossRef]
  132. Sakitani, K.; Nishizawa, T.; Arita, M.; Yoshida, S.; Kataoka, Y.; Ohki, D.; Yamashita, H.; Isomura, Y.; Toyoshima, A.; Watanabe, H.; et al. Early detection of gastric cancer after Helicobacter pylori eradication due to endoscopic surveillance. Helicobacter 2018, 23, e12503. [Google Scholar] [CrossRef]
  133. Take, S.; Mizuno, M.; Ishiki, K.; Kusumoto, C.; Imada, T.; Hamada, F.; Yoshida, T.; Yokota, K.; Mitsuhashi, T.; Okada, H. Risk of gastric cancer in the second decade of follow-up after Helicobacter pylori eradication. J. Gastroenterol. 2020, 55, 281–288. [Google Scholar] [CrossRef]
  134. Mizuno, S.; Miki, I.; Ishida, T.; Yoshida, M.; Onoyama, M.; Azuma, T.; Habu, Y.; Inokuchi, H.; Ozasa, K.; Miki, K.; et al. Prescreening of a High-Risk Group for Gastric Cancer by Serologically Determined Helicobacter pylori Infection and Atrophic Gastritis. Am. J. Dig. Dis. 2010, 55, 3132–3137. [Google Scholar] [CrossRef]
  135. Dang, Y.; Dong, Y.; Mu, Y.; Yan, J.; Lu, M.; Zhu, Y.; Zhang, G. Identification of gastric microbiota biomarker for gastric cancer. Chin. Med. J. 2021, 133, 2765–2767. [Google Scholar] [CrossRef]
  136. Boubrik, F.; Belmouden, A.; El-Kadmiri, N. Potential Non-invasive Biomarkers of Helicobacter pylori—Associated Gastric Cancer. J. Gastrointest. Cancer 2021, 1–8. [Google Scholar] [CrossRef] [PubMed]
  137. Link, A.; Kupcinskas, J. MicroRNAs as non-invasive diagnostic biomarkers for gastric cancer: Current insights and future perspectives. World J. Gastroenterol. 2018, 24, 3313–3329. [Google Scholar] [CrossRef] [PubMed]
  138. Sepulveda, J.L.; Gutierrez-Pajares, J.L.; Luna, A.; Yao, Y.; Tobias, J.W.; Thomas, S.; Woo, Y.; Giorgi, F.; Komissarova, E.V.; Califano, A.; et al. High-definition CpG methylation of novel genes in gastric carcinogenesis identified by next-generation sequencing. Mod. Pathol. Off. J. U. S. Can. Acad. Pathol. Inc. 2016, 29, 182–193. [Google Scholar] [CrossRef] [PubMed]
  139. Sánchez-Zauco, N.; Torres, J.; Gómez, A.; Camorlinga-Ponce, M.; Muñoz-Pérez, L.; Herrera-Goepfert, R.; Medrano-Guzmán, R.; Giono-Cerezo, S.; Maldonado-Bernal, C. Circulating blood levels of IL-6, IFN-γ, and IL-10 as potential diagnostic biomarkers in gastric cancer: A controlled study. BMC Cancer 2017, 17, 384. [Google Scholar]
  140. Xue, Y.; Zhou, L.-Y.; Lu, H.-P.; Liu, J.-Z. Recurrence of Helicobacter pylori infection. Chin. Med. J. 2019, 132, 765–771. [Google Scholar] [CrossRef]
  141. Yee, J.K. Helicobacter pylori colonization of the oral cavity: A milestone discovery. World J. Gastroenterol. 2016, 22, 641–648. [Google Scholar] [CrossRef]
  142. Alkhaldi, N.K.; Alghamdi, W.K.; Alharbi, M.H.; Almutairi, A.S.; Alghamdi, F.T. The Association Between Oral Helicobacter pylori and Gastric Complications: A Comprehensive Review. Cureus 2022, 14, e24703. [Google Scholar] [CrossRef]
  143. Calvet, X.; Ducons, J.; Guardiola, J.; Tito, L.; Andreu, V.; Bory, F.; Guirao, R.; The Group for Eradication Studies from Catalonia and Aragón (Gresca). One-week triple vs. quadruple therapy for Helicobacter pylori infection—A randomized trial. Aliment. Pharmacol. Ther. 2002, 16, 1261–1267. [Google Scholar] [CrossRef]
  144. Katelaris, P.H.; Forbes, G.M.; Talley, N.J.; Crotty, B. A randomized comparison of quadruple and triple therapies for Helicobacter pylori eradication: The QUADRATE study. Gastroenterology 2002, 123, 1763–1769. [Google Scholar] [CrossRef]
  145. Xie, Y.; Pan, X.; Li, Y.; Wang, H.; Du, Y.; Xu, J.; Wang, J.; Zeng, Z.; Chen, Y.; Zhang, G.; et al. 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] [CrossRef]
  146. Luo, M.; Hao, Y.; Tang, M.; Shi, M.; He, F.; Xie, Y.; Chen, W. Application of a social media platform as a patient reminder in the treatment of Helicobacter pylori. Helicobacter 2020, 25, e12682. [Google Scholar] [CrossRef] [PubMed]
  147. Nyssen, O.P.; Vaira, D.; Tepes, B.; Kupcinskas, L.; Bordin, D.; Pérez-Aisa, Á.; Gasbarrini, A.; Castro-Fernández, M.; Bujanda, L.; Garre, A.; et al. Room for Improvement in the Treatment of Helicobacter pylori Infection: Lessons from the European Registry on H. pylori Management (Hp-EuReg). J. Clin. Gastroenterol. 2022, 56, e98–e108. [Google Scholar] [CrossRef] [PubMed]
  148. Murakami, T.T.; Scranton, R.A.; Brown, H.E.; Harris, R.B.; Chen, Z.; Musuku, S.; Oren, E. Management of Helicobacter Pylori in the United States: Results from a national survey of gastroenterology physicians. Prev. Med. 2017, 100, 216–222. [Google Scholar] [CrossRef] [PubMed]
  149. Saxena, K.; Chauhan, N.; Jain, U. Advances in diagnosis of Helicobacter pylori through biosensors: Point of care devices. Anal. Biochem. 2021, 630, 114325. [Google Scholar] [CrossRef]
  150. Prasad, S. Nanobiosensors: The future for diagnosis of disease? Configurations 2014, 3, 1–10. [Google Scholar] [CrossRef]
  151. Pramanik, P.K.D.; Solanki, A.; Debnath, A.; Nayyar, A.; El-Sappagh, S.; Kwak, K.S. Advancing modern healthcare with nanotechnology, nanobiosensors, and internet of nano things: Taxonomies, applications, architecture, and challenges. IEEE Access 2020, 8, 65230–65266. [Google Scholar] [CrossRef]
  152. Yadav, A.K.; Verma, D.; Chaudhary, N.; Kumar, A.; Solanki, P.R. Aptamer based switches: A futuristic approach for Helicobacter pylori detection. Mater. Lett. 2022, 308, 131239. [Google Scholar] [CrossRef]
  153. Safarov, T.; Kıran, 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, 17, 829–840. [Google Scholar] [CrossRef]
  154. Rokkas, T.; Gisbert, J.P.; Malfertheiner, P.; Niv, Y.; Gasbarrini, A.; Leja, M.; Megraud, F.; O’morain, C.; Graham, D.Y. Comparative Effectiveness of Multiple Different First-Line Treatment Regimens for Helicobacter pylori Infection: A Network Meta-analysis. Gastroenterology 2021, 161, 495–507.e494. [Google Scholar] [CrossRef]
  155. Fu, J.; Sun, C.-F.; He, H.-Y.; Ojha, S.C.; Shi, H.; Deng, C.-L.; Sheng, Y.-J. The effect of CYP2C19 gene polymorphism on the eradication rate of Helicobacter pylori by proton pump inhibitors-containing regimens in Asian populations: A meta-analysis. Pharmacogenomics 2021, 22, 859–879. [Google Scholar] [CrossRef]
  156. Zihlif, M.; Bashaireh, B.; Rashid, M.; Almadani, Z.; Jarrar, Y. Effect of major CYP2C19 genetic polymorphisms on Helicobacter pylori eradication based on different treatment regimens. Biomed. Rep. 2022, 16, 1485. [Google Scholar] [CrossRef]
  157. Clyne, M.; Rowland, M. The Role of Host Genetic Polymorphisms in Helicobacter pylori Mediated Disease Outcome. Adv. Exp. Med. Biol. 2019, 1149, 151–172. [Google Scholar] [CrossRef] [PubMed]
  158. Karbalaei, M.; Khorshidi, M.; Sisakht-Pour, B.; Ghazvini, K.; Farsiani, H.; Youssefi, M.; Keikha, M. What are the effects of IL-1β (rs1143634), IL-17A promoter (rs2275913) and TLR4 (rs4986790) gene polymorphism on the outcomes of infection with H. pylori within as Iranian population; A systematic review and meta-analysis. Gene Rep. 2020, 20, 100735. [Google Scholar] [CrossRef]
  159. Eed, E.M.; Hawash, Y.A.; Khalifa, A.S.; Alsharif, K.F.; Alghamdi, S.A.; Almalki, A.A.; Almehmadi, M.M.; Ismail, K.A.; Taha, A.A.; Saber, T. Association of Toll-Like Receptors 2, 4, 9 and 10 Genes Polymorphisms and Helicobacter pylori-Related Gastric Diseases in Saudi Patients. Indian J. Med. Microbiol. 2020, 38, 94–100. [Google Scholar] [CrossRef]
  160. Shinozaki, S.; Kobayashi, Y.; Osawa, H.; Sakamoto, H.; Hayashi, Y.; Lefor, A.K.; Yamamoto, H. Effectiveness and Safety of Vonoprazan versus Proton Pump Inhibitors for Second-Line Helicobacter pylori Eradication Therapy: Systematic Review and Meta-Analysis. Digestion 2021, 102, 319–325. [Google Scholar] [CrossRef]
  161. Lyu, Q.J.; Pu, Q.H.; Zhong, X.F.; Zhang, J. Efficacy and safety of vonoprazan-based versus proton pump inhibitor-based triple therapy for Helicobacter pylori eradication: A meta-analysis of randomized clinical trials. Biomed. Res. Int. 2019, 2019, 9781212. [Google Scholar]
  162. Kiyotoki, S.; Nishikawa, J.; Sakaida, I. Efficacy of Vonoprazan for Helicobacter pylori Eradication. Intern. Med. 2020, 59, 153–161. [Google Scholar] [CrossRef]
  163. Brogden, K.A. Antimicrobial peptides: Pore formers or metabolic inhibitors in bacteria? Nat. Rev. Microbiol. 2005, 3, 238–250. [Google Scholar]
  164. Castano, A.P.; Demidova, T.N.; Hamblin, M.R. Mechanisms in photodynamic therapy: Part two-cellular signaling, cell metabolism and modes of cell death. Photodiagn. Photodyn. Ther. 2005, 2, 1–23. [Google Scholar]
  165. Viertel, T.M.; Ritter, K.; Horz, H.P. Viruses versus bacteria-novel approaches to phage therapy as a tool against multi-drug-resistant pathogens. J. Antimicrob. Chemother. 2014, 69, 2326–2336. [Google Scholar]
  166. Sousa, C.; Ferreira, R.; Azevedo, N.F.; Oleastro, M.; Azeredo, J.; Figueiredo, C.; Melo, L.D.R. Helicobacter pylori infection: From standard to alternative treatment strategies. Crit. Rev. Microbiol. 2022, 48, 376–396. [Google Scholar] [CrossRef]
  167. Sutton, P.; Boag, J.M. Status of vaccine research and development for Helicobacter pylori. Vaccine 2018, 37, 7295–7299. [Google Scholar] [CrossRef] [PubMed]
Table 2. Treatment options for Hp.
Table 2. Treatment options for Hp.
First-Line Treatments
RegimenDosing FrequencyDurationIndicationsNotesStudy
Triple
PPI (variable dose) a
CA (500 mg)
AM b (1 g) OR MZ (500 mg)

BID
BID
BID
14 daysFirst-line treatment in regions where CA resistance is low (<15%) or with high proven local eradication rates (>85%) and in patients with no previous macrolide exposure.A few studies are listed to summarize global resistance rates [14,101].
Eradication rates of up to 92.6% have been reported with triple therapy when potent and long-lasting gastric acid inhibitors, such as K+-competetive blocker vonoprazan, are used [102].
Maastricht
ACG
Toronto
Concomitant
PPI (variable dose) a
CA (500 mg)
AM (1 g)
MZ (500 mg)

BID
BID
BID
BID
10–14 daysFirst-line treatment, especially in regions where CA resistance is high (>15%) and metronidazole resistance is low. Maastricht
ACG
Toronto
Quadruple Bismuth
PPI (variable dose) a
Bismuth (variable dose and preparation) c
AM
MZ


QID
BID
TID or QID
10–14 daysFirst-line treatment, especially in regions where CA and MZ resistances are high. Maastricht
ACG
Toronto
Chinese
Spanish
Second-line treatments
Quadruple Bismuth
PPI (high dose) a
Bismuth (variable dose and preparation) c
TZ (500 mg)
MZ (500 mg)

BID
QID
QID
TID to QID
10–14 daysCan be used as a first-line treatment. Used as a second-line treatment if:
  • Triple or concomitant treatment failed; or
  • Earlier bismuth quadruple treatment failed (two different antibiotics need to be used).
The list of antibiotics that can be used alongside bismuth includes [103]:
Clarithromycin;
Amoxicillin;
Doxycycline;
Nitroimidazole;
Furazolidone; and
Levofloxacin.
Maastricht
ACG
Toronto
Chinese
Spanish
AGA
Levofloxacin Regimens
Levofloxacin (500 mg)
Amoxicillin (1 g)
PPI (high dose) a

QD
BID
BID
10–14 daysPotential first-line treatment in areas with low fluroquinolone resistance (reference to ACG).
Second-line treatment after failure of a bismuth regimen.
Only ACG suggests this as a first-line treatment in regions where levofloxacin resistance is low.
Levofloxacin can be replaced by sitofloxacin.
Maastricht
ACG
Toronto
Chinese
Spanish
AGA
Rescue treatments d
High-dose dual
PPI (high dose) a
AM (750 mg or 1 g)

BID
QID or TID respectively
14 daysSalvage therapy after two eradication failures.Amoxicillin resistance rates are still low globally.ACG
Toronto
AGA
Rifabutin-based triple
Rifabutin
Amoxicillin
PPI (high dose) a

QD
TID
BID
14 daysSalvage therapy after two (Malfertheimer) or three (fallone) eradication failures. AGA guidelines suggest use as a second-line treatment after failed Bismuth therapy.There is some concern about increasing M. tuberculosis resistance as a result of this treatment.Maastricht
Toronto
AGA
Spanish
Alternative or adjunctive treatments
Statins
Atorvastatin (40 mg)
Simvastatin (20 mg)

QD
BID

14 days
Experimental useStatins have been shown to have antibacterial and anti-inflammatory effects [104,105]. One study found that statins reduced Hp burden in macrophages and increased Hp-infected macrophage autophagy [106].AGA
Probiotics e 14 daysExperimental useProbiotic strains have been shown to have a beneficial effect on eradication and to reduce treatment adverse effects, including:
Lactobacillus;
Bifidobacterium;
Lactiplantibacillus; and
Saccharomyces
AGA
Kyoto
Viazis et al., 2022 [107]
Notes: Abbreviations: PPI—proton pump inhibitor; CA—clarithromycin; AM—amoxicillin; MZ—metronidazole; BID—bidaily; QID—quad daily; TID—tridaily; TZ—tetracycline; QD—once daily; a dose varies depending on PPI used. Standard doses include esomeprazole 20 mg, lansoprazole 30 mg, omeprazole 20 mg, pantoprazole 40 mg, and rabeprazole 20 mg. High dose implies double the standard dose. b In patients with a penicillin allergy, amoxicillin should be substituted for metronidazole. c Bismuth can come in multiple preparations; the most common preparations are: Bismuth subsalicylate (262 mg), two tablets QID; colloidal bismuth subcitrate (120 mg), one tablet QID; bismuth biskalcitrate (140 mg), three tablets QID; bismuth subcitrate potassium (140 mg), three tablets QID. d Alternative antibiotics that can be used in rescue treatments include sitafloxacin, tinidazole, and furazolidone. e Lactobacillus and Bifidobacterium are supported by a growing body of evidence, whereas the benefits of Lactiplantibacillus and Saccharomyces are supported by a limited number of studies.
Table 3. Medication adverse events resulting from H. pylori treatment.
Table 3. Medication adverse events resulting from H. pylori treatment.
Adverse EffectReported Frequency *Associated Treatment Group
Taste disturbance/oral mucositis [143,144,145]17–44%Triple therapy
Nausea [143,144]7–31%Bismuth
Diarrhea [143,144,145]7–33%Triple
Dyspepsia [143,145]3–11%
Reduced appetite [143,144]4–12%
Vomiting [144,145]3–6%
Abdominal pain [144,145]8–20%
Headache [144,145]7–31%Bismuth
Rash [144]3–7%Bismuth
Discoloration of feces [144,145]4–16%Bismuth
Oral/vaginal candidiasis [143,145]1–4%
Notes: * frequencies represent a range report in the selected studies comparing different treatment modalities; † not based on statistical comparison between different treatments, rather an observed difference in frequency between different treatment modalities. However, Calvet et al. [143] showed that triple therapy was significantly associated with more frequent taste disturbance.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Shatila, M.; Thomas, A.S. Current and Future Perspectives in the Diagnosis and Management of Helicobacter pylori Infection. J. Clin. Med. 2022, 11, 5086. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm11175086

AMA Style

Shatila M, Thomas AS. Current and Future Perspectives in the Diagnosis and Management of Helicobacter pylori Infection. Journal of Clinical Medicine. 2022; 11(17):5086. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm11175086

Chicago/Turabian Style

Shatila, Malek, and Anusha Shirwaikar Thomas. 2022. "Current and Future Perspectives in the Diagnosis and Management of Helicobacter pylori Infection" Journal of Clinical Medicine 11, no. 17: 5086. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm11175086

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

Article Metrics

Back to TopTop